Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Molecules ; 24(7)2019 Apr 10.
Article in English | MEDLINE | ID: mdl-30974725

ABSTRACT

The scope of this article is to provide a brief general introduction to heparan sulfate (HS) and heparin, and attempt to identify some of the central challenges regarding research into the chemistry and biology of glycosaminoglycans (GAGs), some of which are the subject of contributions to the special issue of Molecules (published in volume 23, 2018) entitled 'Heparan Sulfate and Heparin: Challenges and Controversies' [...].


Subject(s)
Biomedical Research , Heparin , Heparitin Sulfate , Periodicals as Topic
3.
Respir Care ; 61(5): 586-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26732142

ABSTRACT

BACKGROUND: Mucolytic agents, such as nebulized hypertonic saline, may improve airway clearance and shorten the duration of mechanical ventilation, but prospective blinded studies in children undergoing mechanical ventilation are lacking. METHODS: Children <18 y old who had been intubated for <12 h and had an expected duration of mechanical ventilation of >48 additional h were prophylactically given 3 mL of either nebulized hypertonic saline or placebo (0.9% saline) 4 times/d. The primary outcome was duration of mechanical ventilation. Ventilator parameters and the presence of wheezing were recorded before and after study drug administration. RESULTS: The duration of mechanical ventilation was significantly longer in children treated with hypertonic saline (208.1 [interquartile range 136.3-319.8] h) versus those treated with placebo (129.5 [interquartile range 74.4-146.1] h) (P = .03 by Wilcoxon rank-sum test). After adjusting for baseline levels of PEEP, the duration of mechanical ventilation did not differ between groups. Mechanical ventilation parameters, including dead space and dynamic compliance, did not differ between measurements taken before study drug administration versus measurements taken after. New onset wheezing following study drug administration was rare (1.0% with hypertonic saline vs 3.0% with placebo, P = .36 by chi-square test). CONCLUSIONS: Administering prophylactic nebulized hypertonic saline to mechanically ventilated children did not improve clinically relevant outcomes, including duration of mechanical ventilation. Wheezing after hypertonic saline treatment was rare.


Subject(s)
Expectorants/administration & dosage , Nebulizers and Vaporizers , Respiration Disorders/therapy , Respiration, Artificial , Saline Solution, Hypertonic/administration & dosage , Double-Blind Method , Female , Humans , Infant , Male , Pilot Projects , Ventilators, Mechanical
4.
Biophys J ; 104(8): 1720-30, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23601319

ABSTRACT

Fibroblast growth factors (FGFs) utilize cell surface heparan sulfate as a coreceptor in the assembly of signaling complexes with FGF-receptors on the plasma membrane. Here we undertake a complete thermodynamic characterization of the assembly of the FGF signaling complex using isothermal titration calorimetry. Heparin fragments of defined length are used as chemical analogs of the sulfated domains of heparan sulfate and examined for their ability to oligomerize FGF1. Binding is modeled using the McGhee-von Hippel formalism for the cooperative binding of ligands to a monodimensional lattice. Oligomerization of FGFs on heparin is shown to be mediated by positive cooperativity (α = 6). Heparin octasaccharide is the shortest length capable of dimerizing FGF1 and on longer heparin chains FGF1 binds with a minimal footprint of 4.2 saccharide units. The thermodynamics and stoichiometry of the ternary complex suggest that in solution FGF1 binds to heparin in a trans-dimeric manner before FGFR recruitment.


Subject(s)
Fibroblast Growth Factors/metabolism , Heparin/pharmacology , Protein Multimerization , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Amino Acid Sequence , Fibroblast Growth Factors/chemistry , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Protein Binding/drug effects , Protein Structure, Tertiary , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Thermodynamics
5.
Science ; 332(6028): 484-8, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21454754

ABSTRACT

Heparan and chondroitin sulfate proteoglycans (HSPGs and CSPGs, respectively) regulate numerous cell surface signaling events, with typically opposite effects on cell function. CSPGs inhibit nerve regeneration through receptor protein tyrosine phosphatase sigma (RPTPσ). Here we report that RPTPσ acts bimodally in sensory neuron extension, mediating CSPG inhibition and HSPG growth promotion. Crystallographic analyses of a shared HSPG-CSPG binding site reveal a conformational plasticity that can accommodate diverse glycosaminoglycans with comparable affinities. Heparan sulfate and analogs induced RPTPσ ectodomain oligomerization in solution, which was inhibited by chondroitin sulfate. RPTPσ and HSPGs colocalize in puncta on sensory neurons in culture, whereas CSPGs occupy the extracellular matrix. These results lead to a model where proteoglycans can exert opposing effects on neuronal extension by competing to control the oligomerization of a common receptor.


Subject(s)
Axons/physiology , Chondroitin Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Sensory Receptor Cells/physiology , Amino Acid Sequence , Animals , Binding Sites , Cell Membrane/metabolism , Cells, Cultured , Chondroitin Sulfate Proteoglycans/chemistry , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Crystallography, X-Ray , Extracellular Matrix , Ganglia, Spinal , Glypicans/metabolism , Growth Cones/metabolism , Heparan Sulfate Proteoglycans/chemistry , Heparitin Sulfate/analogs & derivatives , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Humans , Mice , Models, Biological , Models, Molecular , Molecular Sequence Data , Neurites/physiology , Neurocan/metabolism , Protein Conformation , Protein Multimerization , Protein Structure, Tertiary
6.
J Biol Chem ; 286(8): 6241-52, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21148566

ABSTRACT

Heparan sulfate proteoglycans (HSPG) encompass some of the most abundant macromolecules on the surface of almost every cell type. Heparan sulfate (HS) chains provide a key interaction surface for the binding of numerous proteins such as growth factors and morphogens, helping to define the ability of a cell to respond selectively to environmental cues. The specificity of HS-protein interactions are governed predominantly by the order and positioning of sulfate groups, with distinct cell types expressing unique sets of HS epitopes. Embryos deficient in HS-synthesis (Ext1(-/-)) exhibit pre-gastrulation lethality and lack recognizable organized mesoderm and extraembryonic tissues. Here we demonstrate that embryonic stem cells (ESCs) derived from Ext1(-/-) embryos are unable to differentiate into hematopoietic lineages, instead retaining ESC marker expression throughout embryoid body (EB) culture. However hematopoietic differentiation can be restored by the addition of soluble heparin. Consistent with specific size and composition requirements for HS:growth factor signaling, chains measuring at least 12 saccharides were required for partial rescue of hematopoiesis with longer chains (18 saccharides or more) required for complete rescue. Critically N- and 6-O-sulfate groups were essential for rescue. Heparin addition restored the activity of multiple signaling pathways including bone morphogenic protein (BMP) with activation of phospho-SMADs re-established by the addition of heparin. Heparin addition to wild-type cultures also altered the outcome of differentiation, promoting hematopoiesis at low concentrations, yet inhibiting blood formation at high concentrations. Thus altering the levels of HS and HS sulfation within differentiating ESC cultures provides an attractive and accessible mechanism for influencing cell fate.


Subject(s)
Anticoagulants/pharmacology , Cell Differentiation/drug effects , Embryonic Stem Cells/metabolism , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Heparin/pharmacology , Heparitin Sulfate/pharmacology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism
7.
Thromb Haemost ; 102(6): 1265-73, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19967160

ABSTRACT

Heparin and its derivatives have been shown to inhibit angiogenesis and metastasis formation. Accordingly, we investigated the effect of heparin fragments containing 4 to 22 monomers on human melanoma cell proliferation, migration and invasion in vitro as well as on the in vivo metastatic potential in a SCID mouse model. Only oligosaccharide dp18 had significant inhibitory effect on cell proliferation. In contrast, cell migration was inhibited by all oligosaccharides studied except dp8 and dp22. Anti-CD44v3 antibody stimulated cell migration and invasion, and this effect could be attenuated by oligosaccharides dp4 and dp18. These fragments also inhibited the catalytic activity of myosin light chain phosphatase as well. Moreover, oligosaccharides dp4 and dp18 reduced the number of lung colonies formed in SCID mice intravenously injected with human melanoma cells, while dp22 proved to be ineffective in this respect. These studies revealed that fragments of heparin have an antimigratory and antimetastatic potential. These fragments lack the haemostatic effect of heparin, suggesting that they are potential specific antimetastatic agents in anticancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Melanoma, Experimental/drug therapy , Oligosaccharides/therapeutic use , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Heparin, Low-Molecular-Weight/chemistry , Humans , Hyaluronan Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Oligosaccharides/chemistry , Protein Phosphatase 1/antagonists & inhibitors , Transplantation, Heterologous
8.
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
9.
Stem Cells ; 26(12): 3108-18, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18787209

ABSTRACT

Heparan sulfate (HS) is a mandatory coreceptor for many growth factors and morphogens involved in embryonic development; its bioactivity is dictated by complex sulfation motifs embedded within the polymer chain. Using a panel of HS-specific antibodies we have identified a unique HS epitope recognized by antibody HS4C3 that is selectively expressed during differentiation of embryonic stem (ES) cells along the mesodermal lineage to the hemangioblast stage. The appearance of this high-affinity HS4C3-binding (HS4C3(high)) epitope is transient; the epitope is specifically expressed within the emerging Brachyury(+) (Bry(+)) population and marks those cells that will become fetal liver kinase 1 (Flk1)(+). Fluorescence-activated cell sorting (FACS) separation and colony forming assays revealed that HS4C3(high)/Flk1(+) cells have a dramatically increased potential to form both blast and endothelial colonies, both of which depend upon the HS-binding growth factor vascular endothelial growth factor. Critically, expression of this HS epitope is tightly regulated, disappearing from the cell surface as the resultant hematopoietic lineages mature, in a similar manner to protein markers Bry and Flk1. In vivo studies showed a remarkable correlation with in vitro findings, with expression of HS4C3-binding epitopes restricted to newly formed mesodermal tissues during gastrulation. We believe this is the first time a defined HS epitope has been implicated in a specific developmental pathway and that this provides, in addition, a novel enrichment technique for the isolation of hemangioblasts from mixed differentiated ES cell cultures.


Subject(s)
Epitopes/chemistry , Gene Expression Regulation, Developmental , Heparitin Sulfate/chemistry , Mesoderm/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Separation , Fetal Proteins/biosynthesis , Flow Cytometry , Hemangioblasts/metabolism , Mice , Models, Biological , T-Box Domain Proteins/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
10.
J Biol Chem ; 283(19): 13001-8, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18281281

ABSTRACT

Heparin-like saccharides play an essential role in binding to both fibroblast growth factors (FGF) and their receptors at the cell surface. In this study we prepared a series of heparin oligosaccharides according to their size and sulfation level. We then investigated their affinity for FGF2 and their ability to support FGF2 mitogenesis of heparan sulfate-deficient cells expressing FGFR1c. Tetra- and hexasaccharides bound FGF2, but failed to dimerize the growth factor. Nevertheless, these saccharides promoted FGF2-mediated cell growth. Furthermore, whereas enzymatic removal of the non-reducing end 2-O-sulfate group had little effect on the 1:1 interaction with FGF2, it eliminated the mitogenic activity of these saccharides. This evidence supports the symmetric two-end model of ternary complex formation. In contrast, even at very low concentrations, octasaccharide and larger heparin fragments conferred a potent mitogenic activity that was independent of terminal 2-O-sulfation. This correlated with the ability to dimerize FGF2 in an apparently cooperative manner. This data suggests that potent mitogenic signaling results from heparin-mediated trans-dimerization of FGF2, consistent with the asymmetric model of ternary complex formation. We propose that, depending on saccharide structure, there are different architectures and modes of ternary complex assembly that differ in stability and/or efficiency of transmembrane signaling.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparin/pharmacology , Mitosis/drug effects , Animals , Cell Line , Chromatography, High Pressure Liquid , Heparin/chemistry , Heparin/isolation & purification , Mice , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Sulfates/chemistry , Sulfates/metabolism
11.
Anal Chem ; 80(23): 9204-12, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19551986

ABSTRACT

Structural elucidation of glycosaminoglycans (GAGs) is one of the major challenges in biochemical analysis. This is mainly because of the diversity of GAG sulfation and N-acetylation patterns and variations in uronate isomers. ESI-MS and recently MALDI-MS methodologies are important strategies for investigating the molecular structure of GAGs. However, the interpretation of MS data produced by these strategies must take into account a large number of variables (including the number of monosaccharide residues, acetylations, sulfate groups, multiple charges, and exchanges between different cations). We have developed a bioinformatics tool to assist this complex interpretation task. The software is based on GlycoWorkbench, a tool for semiautomatic interpretation of glycan MS data. The tool generates the sugar backbones in all their variants (GAG family, composition, acetylation positions, and number of sulfates) and automatically matches them with the selected MS peaks. The backbones corresponding to a given peak are validated against the selected MS/MS peaks by generating all possible fragmentations. Native chondroitin sulfate and heparin oligosaccharides as well as chemically modified heparin oligomers have been successfully analyzed by MALDI- and ESI-MS and MS/MS, and the results of the semiautomated annotation of these mass spectra are presented here.


Subject(s)
Glycosaminoglycans/analysis , Mass Spectrometry/methods , Software , Chondroitin Sulfates/analysis , Chondroitin Sulfates/chemistry , Glycosaminoglycans/chemistry , Heparin/analysis , Heparin/chemistry , Molecular Structure , Oligosaccharides/analysis , Oligosaccharides/chemistry
12.
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
13.
Glycobiology ; 18(1): 28-41, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18006589

ABSTRACT

NKp30 is a natural cytotoxicity receptor expressed by human NK cells and involved in NK lytic activity. We previously published that membranal heparan sulfate serves as a coligand for human NKp30. In the present study, we complement our results by showing direct binding of recombinant NKp30 to immobilized heparin. The heparan sulfate epitope(s) on target tumor cells and the heparin epitope(s) recognized by NKp30 share similar characteristics. Warren and colleagues (Warren HS, Jones AL, Freeman C, Bettadapura J, Parish CR. 2005. Evidence that the cellular ligand for the human NK cell activation receptor NKp30 is not a heparan sulfate glycosaminoglycan. J Immunol. 175:207-212) published that NKp30 does not bind to membranal heparan sulfate on target cells and that heparan sulfate is not involved in NKp30-mediated lysis. In the current study, we examine the binding of six different recombinant NKp30s to membranal heparan sulfate and conclude that NKp30 does interact with membranal heparan sulfate. Yet, two of the six recombinant NKp30s, including the commercially available recombinant NKp30 (employed by Warren et al.) did not show heparan sulfate-dependent binding. We demonstrate that this is due to an altered glycosylation of these two recombinant NKp30s. Upon removal of its N-linked glycans, heparan sulfate-dependent binding to tumor cells and direct binding to heparin were restored. Overall, our results emphasize the importance of proper glycosylation for analysis of NKp30 binding to its ligand and that membranal heparan sulfate could serve as a coligand for NKp30. At the cellular level, soluble heparan sulfate enhanced the secretion of IFNgamma by NK-92 natural killer cells activated with anti-NKp30 monoclonal antibody. We discuss the involvement of heparan sulfate binding to NKp30 in NKp30-mediated activation of NK cells.


Subject(s)
Heparitin Sulfate/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Proteins/metabolism , Animals , Binding Sites , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Glycosylation , HeLa Cells , Humans , Natural Cytotoxicity Triggering Receptor 3 , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Polysaccharides/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
J Biol Chem ; 283(7): 3932-41, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18065761

ABSTRACT

Through its interactions with proteins and proteoglycans, thrombospondin-1 (TSP-1) functions at the interface of the cell membrane and the extracellular matrix to regulate matrix structure and cellular phenotype. We have previously determined the structure of the high affinity heparin-binding domain of TSP-1, designated TSPN-1, in association with the synthetic heparin, Arixtra. To establish that the binding of TSPN-1 to Arixtra is representative of the association with naturally occurring heparins, we have determined the structures of TSPN-1 in complex with heparin oligosaccharides containing eight (dp8) and ten (dp10) subunits, by x-ray crystallography. We have found that dp8 and dp10 bind to TSPN-1 in a manner similar to Arixtra and that dp8 and dp10 induce the formation of trans and cis TSPN-1 dimers, respectively. In silico docking calculations partnered with our crystal structures support the importance of arginine residues in positions 29, 42, and 77 in binding sulfate groups of the dp8 and dp10 forms of heparin. The ability of several TSPN-1 domains to bind to glycosaminoglycans simultaneously probably increases the affinity of binding through multivalent interactions. The formation of cis and trans dimers of the TSPN-1 domain with relatively short segments of heparin further enhances the ability of TSP-1 to participate in high affinity binding to glycosaminoglycans. Dimer formation may also involve TSPN-1 domains from two separate TSP-1 molecules. This association would enable glycosaminoglycans to cluster TSP-1.


Subject(s)
Heparin/chemistry , Thrombospondin 1/metabolism , Chromatography, Gel , Crystallization , Crystallography, X-Ray , Dimerization , Humans , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thrombospondin 1/chemistry
15.
Glycobiology ; 17(9): 972-82, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17623722

ABSTRACT

Glycomics is a developing field that provides structural information on complex populations of glycans isolated from tissues, cells and organs. Strategies employing matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) are central to glycomic analysis. Current MALDI-based glycomic strategies are capable of efficiently analyzing glycoprotein and glycosphingolipid glycomes but little attention has been paid to devising glycomic methodologies suited to the analysis of glycosaminoglycan (GAG) polysaccharides which pose special problems for MALDI analysis because of their high level of sulfation and large size. In this paper, we describe MALDI strategies that have been optimized for the analysis of highly sulfated GAG-derived oligosaccharides. A crystalline matrix norharmane, as well as an ionic liquid 1-methylimidazolium alpha-cyano-4-hydroxycinnamate (ImCHCA), have been used for the analysis of heparin di-, tetra-, hexa- and decasaccharides carrying from 2 to 13 sulfate groups. Information about the maximum number of sulfate groups is obtained using the ionic liquid whereas MALDI-TOF/TOF MS/MS experiments using norharmane allowed the determination of the nature of the glycosidic backbone, and more precise information about the presence and the position in the sequence of N-acetylated residues.


Subject(s)
Glycosaminoglycans/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Cattle , Coumaric Acids/chemistry , Crystallization , Glycoproteins/chemistry , Glycosides/chemistry , Heparin/chemistry , Ions , Mass Spectrometry/methods , Models, Chemical , Oligosaccharides/chemistry , Polysaccharides/chemistry , Sphingolipids/chemistry
16.
Stem Cells ; 25(8): 1913-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17464092

ABSTRACT

Embryonic stem (ES) cells can be cultured in conditions that either maintain pluripotency or allow differentiation to the three embryonic germ layers. Heparan sulfate (HS), a highly polymorphic glycosaminoglycan, is a critical cell surface coreceptor in embryogenesis, and in this paper we describe its structural transition from an unusually low-sulfated variant in ES cells to a more highly sulfated form in fluorescence-activated cell sorting-purified neural progenitor cells. The characteristic domain structure of HS was retained during this transformation. However, qualitative variations in surface sulfation patterns between ES and differentiated cells were revealed using HS epitope-specific antibodies and the HS-binding growth factor fibroblast growth factor 2 (FGF-2). Expression profiles of the HS modification enzymes indicated that both "early" (N-sulfotransferases) and "late" (6O- and 3O-sulfotransferases) sulfotransferases contributed to the alterations in sulfation patterning. An HS-null ES line was used to demonstrate the necessity for HS in neural differentiation. HS is a coreceptor for many of the protein effectors implicated in pluripotency and differentiation (e.g., members of the FGF family, bone morphogenic proteins, and fibronectin). We suggest that the stage-specific activities of these proteins are finely regulated by dynamic changes in sulfation motifs in HS chains. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Green Fluorescent Proteins/metabolism , Heparitin Sulfate/chemistry , High Mobility Group Proteins/metabolism , Neurons/cytology , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Heparitin Sulfate/metabolism , Heparitin Sulfate/physiology , High Mobility Group Proteins/genetics , Mice , Mice, Knockout , Neurons/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors , Sulfates/metabolism , Transfection , Tubulin/metabolism
17.
J Biol Chem ; 282(5): 3221-30, 2007 Feb 02.
Article in English | MEDLINE | ID: mdl-17130131

ABSTRACT

Cell surface heparan sulfate (HS) proteoglycans are required in development and postnatal repair. Important classes of ligands for HS include growth factors and extracellular matrix macromolecules. For example, the focal adhesion component syndecan-4 interacts with the III(12-14) region of fibronectin (HepII domain) through its HS chains. The fine structure of HS is critical to growth factor responses, and whether this extends to matrix ligands is unknown but is suggested from in vitro experiments. Cell attachment to HepII showed that heparin oligosaccharides of >or=14 sugar residues were required for optimal inhibition. The presence of N-sulfated glucosamine in the HS was essential, whereas 2-O-sulfation of uronic acid or 6-O-sulfation of glucosamine had marginal effects. In the more complex response of focal adhesion formation through syndecan-4, N-sulfates were again required and also glucosamine 6-O-sulfate. The significance of polymer N-sulfation and sulfated domains in HS was confirmed by studies with mutant Chinese hamster ovary cells where heparan sulfation was compromised. Finally, focal adhesion formation was absent in fibroblasts synthesizing short HS chains resulting from a gene trap mutation in one of the two major glucosaminoglycan polymerases (EXT1). Several separate, specific properties of cell surface HS are therefore required in cell adhesion responses to the fibronectin HepII domain.


Subject(s)
Cell Adhesion/physiology , Fibronectins/chemistry , Fibronectins/metabolism , Heparin/metabolism , Heparitin Sulfate/metabolism , Animals , Binding Sites , Cells, Cultured , Fibroblasts/physiology , Mice , Mice, Knockout , Rats , Recombinant Proteins/metabolism , Syndecan-4/deficiency , Syndecan-4/genetics , Syndecan-4/physiology
18.
Biochem J ; 400(1): 63-73, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16901266

ABSTRACT

HS (heparan sulfate) is essential for normal embryonic development. This requirement is due to the obligatory role for HS in the signalling pathways of many growth factors and morphogens that bind to sulfated domains in the HS polymer chain. The sulfation patterning of HS is determined by a complex interplay of Golgi-located N- and O-sulfotransferases which sulfate the heparan precursor and cell surface endosulfatases that selectively remove 6-O-sulfates from mature HS chains. In the present study we generated single or double knock-out mice for the two murine endosulfatases mSulf1 and mSulf2. Detailed structural analysis of HS from mSulf1-/- fibroblasts showed a striking increase in 6-O-sulfation, which was not seen in mSulf2-/- HS. Intriguingly, the level of 6-O-sulfation in the double mSulf1-/-/2-/- HS was significantly higher than that observed in the mSulf1-/- counterpart. These data imply that mSulf1 and mSulf2 are functionally co-operative. Unlike their avian orthologues, mammalian Sulf activities are not restricted to the highly sulfated S-domains of HS. Mitogenesis assays with FGF2 (fibroblast growth factor 2) revealed that Sulf activity decreases the activating potential of newly-synthesized HS, suggesting an important role for these enzymes in cell growth regulation in embryonic and adult tissues.


Subject(s)
Heparitin Sulfate/metabolism , Sulfatases/metabolism , Sulfotransferases/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Disaccharides/analysis , Epitopes/immunology , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Genotype , Heparitin Sulfate/chemistry , Heparitin Sulfate/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Molecular Structure , Oligosaccharides/analysis , Sulfatases/genetics , Sulfates/metabolism , Sulfotransferases/genetics
19.
J Biol Chem ; 281(21): 14813-22, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16481316

ABSTRACT

Endostatin has attracted considerable attention because of its ability to inhibit angiogenesis. This property of monomeric endostatin contrasts with that of the trimeric endostatin moiety generated from the intact C-terminal domain of collagen XVIII that induces a promigratory phenotype in endothelial cells. This activity is inhibited by monomeric endostatin. In this study we demonstrate that the effect of oligomeric endostatin can also be inhibited by exogenous glycosaminoglycans in a size-dependent manner, with heparin oligosaccharides containing more than 20 monosaccharide residues having optimal inhibitory activity. Oligomeric endostatin was also found to induce morphological changes in Chinese hamster ovary cells, an epithelial cell line. This novel observation allowed the utilization of a panel of Chinese hamster ovary cell mutants with defined glycosaminoglycan biosynthetic defects. The action of oligomeric endostatin on these cells was shown to be dependent on cell surface glycosaminoglycans, principally heparan sulfate with N- and 6-O-sulfation of glucosamine residues rather than iduronate 2-O-sulfation being important for bioactivity. The responsiveness of a cell line (pgsE-606) with globally reduced heparan sulfate sulfation and shortened S domains, however, indicates that overall heparan sulfate domain patterning is the key determinant of the bioactivity of oligomeric endostatin. Purified heparin-monomeric endostatin constructs generated by zero-length cross-linking techniques were found to be unable to inhibit the action of oligomeric endostatin. This indicates a mechanism for the perturbation of oligomeric endostatin action by its monomeric counterpart via competition for glycosaminoglycan attachment sites at the cell surface.


Subject(s)
Cell Membrane/metabolism , Endostatins/chemistry , Heparitin Sulfate/chemistry , Animals , Aorta/cytology , CHO Cells , Cattle , Collagen/chemistry , Cricetinae , Glucosamine/chemistry , Glycosaminoglycans/chemistry , Heparin/chemistry , Iduronic Acid/chemistry , Protein Structure, Tertiary
20.
Biochem J ; 393(Pt 3): 741-8, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16223363

ABSTRACT

The minimal signalling unit for tyrosine kinase receptors is two protomers dimerized by one or more ligands. However, it is clear that maximal signalling requires the formation of larger complexes of many receptors at discrete foci on the cell surface. The biological interactions that lead to this are likely to be diverse and have system specific components. In the present study, we demonstrate that, in the FGF (fibroblast growth factor)-FGFR (FGF receptor) system, multimers of the minimal complex composed of two FGF1 and two FGFR2 protomers can form on a single chain of the co-receptor heparin. Using size-exclusion chromatography, we show that two complexes can form on heparin chains as small as 16 saccharide units. We also show by MS that discrete complexes containing exactly two copies of the minimal signalling unit are formed. However, the doublet of complexes appears to be less co-operative than the formation of the 2:2:1 FGF1:FGFR2:heparin complex, suggesting that this mechanism is one of a number of weaker interactions that might be involved in the formation of a focal complex on the cell surface.


Subject(s)
Fibroblast Growth Factor 1/metabolism , Heparin/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , Fibroblast Growth Factor 1/chemistry , Heparin/chemistry , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Molecular Weight , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Receptor, Fibroblast Growth Factor, Type 2/chemistry , Swine , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL
...