Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Glycobiology ; 31(10): 1319-1329, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34192316

ABSTRACT

Mucopolysaccharidosis type I (MPS-I) is a rare lysosomal storage disorder caused by deficiency of the enzyme alpha-L-iduronidase, which removes iduronic acid in both chondroitin/dermatan sulfate (CS/DS) and heparan sulfate (HS) and thereby contributes to the catabolism of glycosaminoglycans (GAGs). To ameliorate this genetic defect, the patients are currently treated by enzyme replacement and bone marrow transplantation, which have a number of drawbacks. This study was designed to develop an alternative treatment by inhibition of iduronic acid formation. By screening the Prestwick drug library, we identified ebselen as a potent inhibitor of enzymes that produce iduronic acid in CS/DS and HS. Ebselen efficiently inhibited iduronic acid formation during CS/DS synthesis in cultured fibroblasts. Treatment of MPS-I fibroblasts with ebselen not only reduced accumulation of CS/DS but also promoted GAG degradation. In early Xenopus embryos, this drug phenocopied the effect of downregulation of DS-epimerase 1, the main enzyme responsible for iduronic production in CS/DS, suggesting that ebselen inhibits iduronic acid production in vivo. However, ebselen failed to ameliorate the CS/DS and GAG burden in MPS-I mice. Nevertheless, the results propose a potential of iduronic acid substrate reduction therapy for MPS-I patients.


Subject(s)
Fibroblasts/drug effects , Glycosaminoglycans/antagonists & inhibitors , Iduronic Acid/antagonists & inhibitors , Isoindoles/pharmacology , Mucopolysaccharidosis I/drug therapy , Organoselenium Compounds/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Fibroblasts/pathology , Glycosaminoglycans/metabolism , HEK293 Cells , Humans , Iduronic Acid/metabolism , Isoindoles/chemistry , Molecular Structure , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Organoselenium Compounds/chemistry , Structure-Activity Relationship
2.
FASEB J ; 33(11): 11973-11992, 2019 11.
Article in English | MEDLINE | ID: mdl-31398290

ABSTRACT

Invasive spread of glioblastoma (GBM) is linked to changes in chondroitin sulfate (CS) proteoglycan (CSPG)-associated sulfated glycosaminoglycans (GAGs) that are selectively up-regulated in the tumor microenvironment (TME). We hypothesized that inhibiting CS-GAG signaling in the TME would stem GBM invasion. Rat F98 GBM cells demonstrated enhanced preferential cell invasion into oversulfated 3-dimensional composite of CS-A and CS-E [4- and 4,6-sulfated CS-GAG (COMP)] matrices compared with monosulfated (4-sulfated) and unsulfated hyaluronic acid matrices in microfluidics-based choice assays, which is likely influenced by differential GAG receptor binding specificities. Both F98 and human patient-derived glioma stem cells (GSCs) demonstrated a high degree of colocalization of the GSC marker CD133 and CSPGs. The small molecule sulfated GAG antagonist bis-2-methyl-4-amino-quinolyl-6-carbamide (surfen) reduced invasion and focal adhesions in F98 cells encapsulated in COMP matrices and blocked CD133 and antichondroitin sulfate antibody (CS-56) detection of respective antigens in F98 cells and human GSCs. Surfen-treated F98 cells down-regulated CSPG-binding receptor transcripts and protein, as well as total and activated ERK and protein kinase B. Lastly, rats induced with frontal lobe tumors and treated with a single intratumoral dose of surfen demonstrated reduced tumor burden and spread compared with untreated controls. These results present a first demonstration of surfen as an inhibitor of sulfated GAG signaling to stem GBM invasion.-Logun, M. T., Wynens, K. E., Simchick, G., Zhao, W., Mao, L., Zhao, Q., Mukherjee, S., Brat, D. J., Karumbaiah, L. Surfen-mediated blockade of extratumoral chondroitin sulfate glycosaminoglycans inhibits glioblastoma invasion.


Subject(s)
Cell Movement/drug effects , Chondroitin Sulfates/antagonists & inhibitors , Glioblastoma/metabolism , Neoplastic Stem Cells/drug effects , Tumor Microenvironment/drug effects , Urea/analogs & derivatives , AC133 Antigen/metabolism , Animals , Cell Line, Tumor , Chondroitin Sulfates/metabolism , Glioblastoma/pathology , Glioma/metabolism , Glioma/pathology , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/metabolism , Humans , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Rats , Signal Transduction/drug effects , Urea/pharmacology
3.
Int J Mol Sci ; 21(24)2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33333756

ABSTRACT

Orthodontic tooth movement (OTM) creates compressive and tensile strain in the periodontal ligament, causing circulation disorders. Hypoxia-inducible factor 1α (HIF-1α) has been shown to be primarily stabilised by compression, but not hypoxia in periodontal ligament fibroblasts (PDLF) during mechanical strain, which are key regulators of OTM. This study aimed to elucidate the role of heparan sulfate integrin interaction and downstream kinase phosphorylation for HIF-1α stabilisation under compressive and tensile strain and to which extent downstream synthesis of VEGF and prostaglandins is HIF-1α-dependent in a model of simulated OTM in PDLF. PDLF were subjected to compressive or tensile strain for 48 h. In various setups HIF-1α was experimentally stabilised (DMOG) or destabilised (YC-1) and mechanotransduction was inhibited by surfen and genistein. We found that HIF-1α was not stabilised by tensile, but rather by compressive strain. HIF-1α stabilisation had an inductive effect on prostaglandin and VEGF synthesis. As expected, HIF-1α destabilisation reduced VEGF expression, whereas prostaglandin synthesis was increased. Inhibition of integrin mechanotransduction via surfen or genistein prevented stabilisation of HIF-1α. A decrease in VEGF expression was observed, but not in prostaglandin synthesis. Stabilisation of HIF-1α via integrin mechanotransduction and downstream phosphorylation of kinases seems to be essential for the induction of VEGF, but not prostaglandin synthesis by PDLF during compressive (but not tensile) orthodontic strain.


Subject(s)
Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mechanotransduction, Cellular , Periodontal Ligament/metabolism , Adolescent , Adult , Cells, Cultured , Female , Fibroblasts/drug effects , Focal Adhesion Kinase 1/antagonists & inhibitors , Genistein/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Glycosaminoglycans/antagonists & inhibitors , Humans , Indazoles/pharmacology , Integrins/antagonists & inhibitors , Male , Mechanotransduction, Cellular/drug effects , Mechanotransduction, Cellular/genetics , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Phosphorylation , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/biosynthesis , Prostaglandins/metabolism , Protein Stability/drug effects , Signal Transduction/drug effects , Stress, Mechanical , Tooth Movement Techniques , Urea/analogs & derivatives , Urea/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Arterioscler Thromb Vasc Biol ; 38(6): 1258-1270, 2018 06.
Article in English | MEDLINE | ID: mdl-29674476

ABSTRACT

The glycosaminoglycans (GAGs) heparan sulfate, dermatan sulfate, and heparin are important anticoagulants that inhibit clot formation through interactions with antithrombin and heparin cofactor II. Unfractionated heparin, low-molecular-weight heparin, and heparin-derived drugs are often the main treatments used clinically to handle coagulatory disorders. A wide range of proteins have been reported to bind and neutralize these GAGs to promote clot formation. Such neutralizing proteins are involved in a variety of other physiological processes, including inflammation, transport, and signaling. It is clear that these interactions are important for the control of normal coagulation and influence the efficacy of heparin and heparin-based therapeutics. In addition to neutralization, the anticoagulant activities of GAGs may also be regulated through reduced synthesis or by degradation. In this review, we describe GAG neutralization, the proteins involved, and the molecular processes that contribute to the regulation of anticoagulant GAG activity.


Subject(s)
Anticoagulants/therapeutic use , Blood Coagulation/drug effects , Glycosaminoglycans/antagonists & inhibitors , Heparin Antagonists/therapeutic use , Heparin/therapeutic use , Animals , Anticoagulants/adverse effects , Binding Sites , Dermatan Sulfate/antagonists & inhibitors , Dermatan Sulfate/blood , Glycosaminoglycans/blood , Heparin/adverse effects , Heparin Antagonists/adverse effects , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/blood , Humans , Protein Binding
5.
Int J Mol Sci ; 20(2)2019 Jan 14.
Article in English | MEDLINE | ID: mdl-30646511

ABSTRACT

Obstacles to effective therapies for mucopolysaccharidoses (MPSs) determine the need for continuous studies in order to enhance therapeutic strategies. Dimethyl sulfoxide (DMSO) is frequently utilised as a solvent in biological studies, and as a vehicle for drug therapy and the in vivo administration of water-insoluble substances. In the light of the uncertainty on the mechanisms of DMSO impact on metabolism of glycosaminoglycans (GAGs) pathologically accumulated in MPSs, in this work, we made an attempt to investigate and resolve the question of the nature of GAG level modulation by DMSO, the isoflavone genistein solvent employed previously by our group in MPS treatment. In this work, we first found the cytotoxic effect of DMSO on human fibroblasts at concentrations above 3%. Also, our results displayed the potential role of DMSO in the regulation of biological processes at the transcriptional level, then demonstrated a moderate impact of the solvent on GAG synthesis. Interestingly, alterations of lysosomal ultrastructure upon DMSO treatment were visible. As there is growing evidence in the literature that DMSO can affect cellular pathways leading to numerous changes, it is important to expand our knowledge concerning this issue.


Subject(s)
Dimethyl Sulfoxide/administration & dosage , Genistein/administration & dosage , Lysosomal Storage Diseases/drug therapy , Mucopolysaccharidoses/drug therapy , Cell Line , Cell Proliferation/drug effects , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Glycosaminoglycans/antagonists & inhibitors , Humans , Isoflavones/metabolism , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Lysosomes/drug effects , Lysosomes/ultrastructure , Mucopolysaccharidoses/metabolism , Mucopolysaccharidoses/pathology
6.
Anal Chem ; 90(8): 5201-5208, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29533603

ABSTRACT

The structures and amounts of glycosaminoglycan (GAG) produced by cells have attracted much interest because GAG biosynthesis activity can change in cellular processes such as disease and differentiation. ß-Xylosides, also called saccharide primers, have been used as artificial acceptors not only to generate GAG oligosaccharides in cells and tissues but also to investigate their biosynthetic pathways. Various analytical methods have been applied to confirm the structure and amounts of GAG oligosaccharides elongated using saccharide primers, yet sample preparation processes such as solid-phase extraction in analysis can cause experimental error and disrupt accurate comparative quantification of glycosylated products. In this study, we developed a new quantification method using a deuterium-labeled saccharide primer. The "heavy" and "light" primers were chemically synthesized, and priming abilities were confirmed by liquid chromatography-tandem mass spectrometry. Relative peak areas of light/heavy products showed good linearity and were well correlated with the theoretical amounts of glycosylated products. Then, as a validation study, we carried out a biosynthesis inhibition assay using known GAG biosynthesis inhibitors. According to the relative quantification using saccharide primers, differences in the mode-of-action among the four GAG biosynthesis inhibitors were dependent on the GAG biosynthetic pathway. Our results indicate that the method will likely forge a new path for comparative glycosaminoglycomics using cultured cells and tissues.


Subject(s)
Glycosaminoglycans/analysis , Glycosides/chemistry , Isotope Labeling , Oligosaccharides/chemistry , Azaserine/pharmacology , Brefeldin A/pharmacology , Cell Line , Genistein/pharmacology , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/biosynthesis , Glycosylation , Humans , Molecular Structure , Rhodamines/pharmacology
7.
Org Biomol Chem ; 15(27): 5656-5668, 2017 Jul 21.
Article in English | MEDLINE | ID: mdl-28653068

ABSTRACT

Heparin and heparan sulfate glycosaminoglycans are long, linear polysaccharides that are made up of alternating dissacharide sequences of sulfated uronic acid and amino sugars. Unlike heparin, which is only found in mast cells, heparan sulfate is ubiquitously expressed on the cell surface and in the extracellular matrix of all animal cells. These negatively-charged glycans play essential roles in important cellular functions such as cell growth, adhesion, angiogenesis, and blood coagulation. These biomolecules are also involved in pathophysiological conditions such as pathogen infection and human disease. This review discusses past and current methods for targeting these complex biomolecules as a novel therapeutic strategy to treating disorders such as cancer, neurodegenerative diseases, and infection.


Subject(s)
Glycosaminoglycans/antagonists & inhibitors , Heparin/metabolism , Heparitin Sulfate/antagonists & inhibitors , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Animals , Glycosaminoglycans/chemistry , Heparin/chemistry , Heparitin Sulfate/chemistry , Humans , Infections/drug therapy , Infections/metabolism , Neoplasms/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Small Molecule Libraries/chemistry
8.
FASEB J ; 29(7): 2993-3002, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25868729

ABSTRACT

Glycosaminoglycan (GAG) polysaccharides have been implicated in a variety of cellular processes, and alterations in their amount and structure have been associated with diseases such as cancer. In this study, we probed 11 sugar analogs for their capacity to interfere with GAG biosynthesis. One analog, with a modification not directly involved in the glycosidic bond formation, 6F-N-acetyl-d-galactosamine (GalNAc) (Ac3), was selected for further study on its metabolic and biologic effect. Treatment of human ovarian carcinoma cells with 50 µM 6F-GalNAc (Ac3) inhibited biosynthesis of GAGs (chondroitin/dermatan sulfate by ∼50-60%, heparan sulfate by ∼35%), N-acetyl-d-glucosamine (GlcNAc)/GalNAc containing glycans recognized by the lectins Datura stramonium and peanut agglutinin (by ∼74 and ∼43%, respectively), and O-GlcNAc protein modification. With respect to function, 6F-GalNAc (Ac3) treatment inhibited growth factor signaling and reduced in vivo angiogenesis by ∼33%. Although the analog was readily transformed in cells into the uridine 5'-diphosphate (UDP)-activated form, it was not incorporated into GAGs. Rather, it strongly reduced cellular UDP-GalNAc and UDP-GlcNAc pools. Together with data from the literature, these findings indicate that nucleotide sugar depletion without incorporation is a common mechanism of sugar analogs for inhibiting GAG/glycan biosynthesis.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Glycosaminoglycans/biosynthesis , Acetylgalactosamine/chemistry , Acetylgalactosamine/pharmacology , Animals , Cell Line , Chick Embryo , Fibroblast Growth Factor 2/metabolism , Glycosaminoglycans/antagonists & inhibitors , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Physiologic/drug effects , Polysaccharides/antagonists & inhibitors , Polysaccharides/biosynthesis , Signal Transduction/drug effects , Structure-Activity Relationship , Uridine Diphosphate N-Acetylgalactosamine/metabolism , Uridine Diphosphate N-Acetylglucosamine/metabolism , Vascular Endothelial Growth Factor A/metabolism
9.
Cancer Metastasis Rev ; 33(4): 1059-79, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25324146

ABSTRACT

The properties and behavior of tumor cells are closely regulated by their microenvironment. Accordingly, stromal cells and extracellular matrix components can have a pronounced effect on cancer initiation, growth, and progression. The linear glycosaminoglycan hyaluronan (HA) is a major component of the extracellular matrix. Altered synthesis and degradation of HA in the tumor context has been implicated in many aspects of tumor biology. In particular, the accumulation of small HA oligosaccharides (sHA) in the tumor interstitial space may play a decisive role, due to the ability of sHA to activate a number of biological processes that are not modulated by high molecular weight (HMW)-HA. In this article, we review the normal physiological role and metabolism of HA and then survey the evidence implicating HA in tumor growth and progression, focusing in particular on the potential contribution of sHA to these processes.


Subject(s)
Carcinogenesis/genetics , Glycosaminoglycans/metabolism , Hyaluronic Acid/metabolism , Neoplasms/genetics , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/chemistry , Humans , Hyaluronic Acid/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Oligosaccharides/metabolism , Tumor Microenvironment/drug effects
11.
Arterioscler Thromb Vasc Biol ; 32(3): 595-604, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22267481

ABSTRACT

OBJECTIVE: Subendothelial retention of proatherogenic lipoproteins by proteoglycans is critical in atherosclerosis. The aim of this study was to characterize the recognition and antiatherogenic properties of a chimeric monoclonal antibody (mAb) that reacts with sulfated molecules. METHODS AND RESULTS: chP3R99 mAb recognized sulfated glycosaminoglycans, mainly chondroitin sulfate (CS), by ELISA. This mAb blocked ≈70% of low-density lipoprotein (LDL)-CS association and ≈80% of LDL oxidation in vitro, and when intravenously injected to Sprague-Dawley rats (n=6, 1 mg/animal), it inhibited LDL (4 mg/kg intraperitoneally, 1 hour later) retention and oxidation in the artery wall. Moreover, subcutaneous immunization of New Zealand White rabbits (n=19) with chP3R99 mAb (100 µg, 3 doses at weekly intervals) prevented Lipofundin-induced atherosclerosis (2 mL/kg, 8 days) with a 22-fold reduction in the intima-media ratio (P<0.01). Histopathologic and ultrastructural studies showed no intimal alterations or slight thickening, with preserved junctions between endothelial cells and scarce collagen fibers and glycosaminoglycans. In addition, immunization with chP3R99 mAb suppressed macrophage infiltration in aorta and preserved redox status. The atheroprotective effect was associated with the induction of anti-CS antibodies in chP3R99-immunized rabbits, capable of blocking CS-LDL binding and LDL oxidation. CONCLUSION: These results support the use of anti-sulfated glycosaminoglycan antibody-based immunotherapy as a potential tool to prevent atherosclerosis.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Atherosclerosis/prevention & control , Chondroitin Sulfates/antagonists & inhibitors , Glycosaminoglycans/antagonists & inhibitors , Immunization , Animals , Antibody Specificity , Atherosclerosis/chemically induced , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biological Transport , Cell Line , Chondroitin Sulfates/immunology , Disease Models, Animal , Drug Combinations , Enzyme-Linked Immunosorbent Assay , Foam Cells/immunology , Foam Cells/metabolism , Glycosaminoglycans/immunology , Lipoproteins, LDL/metabolism , Mice , Oxidation-Reduction , Oxidative Stress , Phospholipids , Rabbits , Rats , Rats, Sprague-Dawley , Sorbitol
12.
J Virol ; 85(9): 4386-98, 2011 May.
Article in English | MEDLINE | ID: mdl-21307190

ABSTRACT

Herpes simplex virus 1 (HSV-1) is a common human pathogen that causes lifelong latent infection of sensory neurons. Non-nucleoside inhibitors that can limit HSV-1 recurrence are particularly useful in treating immunocompromised individuals or cases of emerging acyclovir-resistant strains of herpesvirus. We report that chebulagic acid (CHLA) and punicalagin (PUG), two hydrolyzable tannins isolated from the dried fruits of Terminalia chebula Retz. (Combretaceae), inhibit HSV-1 entry at noncytotoxic doses in A549 human lung cells. Experiments revealed that both tannins targeted and inactivated HSV-1 viral particles and could prevent binding, penetration, and cell-to-cell spread, as well as secondary infection. The antiviral effect from either of the tannins was not associated with induction of type I interferon-mediated responses, nor was pretreatment of the host cell protective against HSV-1. Their inhibitory activities targeted HSV-1 glycoproteins since both natural compounds were able to block polykaryocyte formation mediated by expression of recombinant viral glycoproteins involved in attachment and membrane fusion. Our results indicated that CHLA and PUG blocked interactions between cell surface glycosaminoglycans and HSV-1 glycoproteins. Furthermore, the antiviral activities from the two tannins were significantly diminished in mutant cell lines unable to produce heparan sulfate and chondroitin sulfate and could be rescued upon reconstitution of heparan sulfate biosynthesis. We suggest that the hydrolyzable tannins CHLA and PUG may be useful as competitors for glycosaminoglycans in the management of HSV-1 infections and that they may help reduce the risk for development of viral drug resistance during therapy with nucleoside analogues.


Subject(s)
Antiviral Agents/metabolism , Glycoproteins/antagonists & inhibitors , Glycosaminoglycans/antagonists & inhibitors , Herpesvirus 1, Human/drug effects , Hydrolyzable Tannins/metabolism , Viral Proteins/antagonists & inhibitors , Virus Internalization/drug effects , Animals , Antiviral Agents/isolation & purification , Benzopyrans/isolation & purification , Benzopyrans/metabolism , Cell Line , Chlorocebus aethiops , Glucosides/isolation & purification , Glucosides/metabolism , Herpesvirus 1, Human/physiology , Humans , Hydrolyzable Tannins/isolation & purification , Microbial Sensitivity Tests , Terminalia/chemistry , Viral Plaque Assay , Virus Inactivation
13.
Orv Hetil ; 153(43): 1719-22, 2012 Oct 28.
Article in Hungarian | MEDLINE | ID: mdl-23089171

ABSTRACT

Pretibial myxoedema is a rare symptom of Graves' disease. Histological studies detected mucopolysaccharide and glycosaminoglycan accumulation, and the role of anti-TSH receptor antibodies has been suggested. In this paper the authors present the case of a 34-year-old male patient with pretibial myxoedema treated successfully with pentoxifylline. In his case history multiple autoimmune diseases (type 1 diabetes mellitus, Graves' disease with severe ophthalmopathy) concomitantly occurred. His severe pretibial myxoedema was undiagnosed and untreated at the time of admission. Because of his diabetes, steroid was contraindicated, which made the choice of the treatment more difficult. He received first intradermal, then intravenous and, finally, oral pentoxifylline, which resulted in a regression of the dermatological symptoms. The beneficial effect of pentoxifylline might be explained by its inhibitory effect of proinflammatory cytokines and proliferation of fibroblasts, and the production of glycosaminoglycan. It was concluded that pentoxifylline can be an effective and safe treatment of pretibial myxoedema.


Subject(s)
Leg Dermatoses/drug therapy , Myxedema/drug therapy , Pentoxifylline/therapeutic use , Administration, Oral , Adult , Autoimmune Diseases/complications , Cell Proliferation/drug effects , Cytokines/antagonists & inhibitors , Fibroblasts/drug effects , Glycosaminoglycans/antagonists & inhibitors , Humans , Infusions, Intravenous , Injections, Intradermal , Leg Dermatoses/metabolism , Leg Dermatoses/pathology , Male , Myxedema/metabolism , Myxedema/pathology , Pentoxifylline/administration & dosage , Phosphodiesterase Inhibitors/therapeutic use , Regional Blood Flow/drug effects , Remission Induction
14.
J Immunol ; 183(5): 3150-9, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19667083

ABSTRACT

Although smallpox was eradicated as a global illness more than 30 years ago, variola virus and other related pathogenic poxviruses, such as monkeypox, remain potential bioterrorist weapons or could re-emerge as natural infections. Poxviruses express virulence factors that down-modulate the host's immune system. We previously compared functional profiles of the poxviral complement inhibitors of smallpox, vaccinia, and monkeypox known as SPICE, VCP (or VICE), and MOPICE, respectively. SPICE was the most potent regulator of human complement and attached to cells via glycosaminoglycans. The major goals of the present study were to further characterize the complement regulatory and heparin binding sites of SPICE and to evaluate a mAb that abrogates its function. Using substitution mutagenesis, we established that (1) elimination of the three heparin binding sites severely decreases but does not eliminate glycosaminoglycan binding, (2) there is a hierarchy of activity for heparin binding among the three sites, and (3) complement regulatory sites overlap with each of the three heparin binding motifs. By creating chimeras with interchanges of SPICE and VCP residues, a combination of two SPICE amino acids (H77 plus K120) enhances VCP activity approximately 200-fold. Also, SPICE residue L131 is critical for both complement regulatory function and accounts for the electrophoretic differences between SPICE and VCP. An evolutionary history for these structure-function adaptations of SPICE is proposed. Finally, we identified and characterized a mAb that inhibits the complement regulatory activity of SPICE, MOPICE, and VCP and thus could be used as a therapeutic agent.


Subject(s)
Complement Activating Enzymes/antagonists & inhibitors , Complement Activating Enzymes/metabolism , Variola virus/immunology , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/physiology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Binding Sites/genetics , Binding Sites/immunology , Binding Sites, Antibody , CHO Cells , Complement Activating Enzymes/genetics , Complement C3b/metabolism , Cricetinae , Cricetulus , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/metabolism , Heparin/metabolism , Humans , Hybridomas , Mice , Molecular Sequence Data , Point Mutation , Variola virus/genetics , Variola virus/pathogenicity , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Virulence Factors/antagonists & inhibitors , Virulence Factors/genetics , Virulence Factors/physiology
15.
Proc Natl Acad Sci U S A ; 105(24): 8375-80, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18544647

ABSTRACT

Glycosaminoglycan storage begins in prenatal life in patients with mucopolysaccharidosis (MPS). In fact, prenatal hydrops is a common manifestation of MPS VII because of beta-glucuronidase (GUS) deficiency. One way to address prenatal storage might be to deliver the missing enzyme across the placenta into the fetal circulation. Maternal IgG is transported across the placenta by the neonatal Fc receptor (FcRn), which recognizes the Fc domain of IgG and mediates transcytosis from maternal to fetal circulation. We hypothesized that we could exploit this process to deliver corrective enzyme to the fetus. To test this hypothesis, the C-terminal fusion protein, GUS-Fc, was compared with native, untagged, recombinant GUS for clearance from the maternal circulation, delivery to the fetus, and reduction of lysosomal storage in offspring of MPS VII mice. We observed that GUS-Fc, infused into pregnant mothers on embryonic days 17 and 18, was transported across the placenta. Similarly infused untagged GUS was not delivered to the fetus. GUS-Fc plasma enzyme activity in newborn MPS VII mice was 1,000 times that seen after administration of untagged GUS and approximately 100 times that of untreated WT newborns. Reduced lysosomal storage in heart valves, liver, and spleen provided evidence that in utero enzyme replacement therapy with GUS-Fc targeted sites of storage in the MPS VII fetus. We hypothesize that this noninvasive approach could deliver the missing lysosomal enzyme to a fetus with any lysosomal storage disease. It might also provide a method for inducing immune tolerance to the missing enzyme or another foreign protein.


Subject(s)
Glucuronidase/therapeutic use , Mucopolysaccharidosis VII/prevention & control , Placenta/metabolism , Receptors, Fc/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Uterus , Animals , Female , Glucuronidase/administration & dosage , Glucuronidase/pharmacokinetics , Glycosaminoglycans/antagonists & inhibitors , Infusions, Parenteral , Lysosomes/metabolism , Mice , Pregnancy , Receptors, Fc/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution
16.
BMC Musculoskelet Disord ; 12: 279, 2011 Dec 12.
Article in English | MEDLINE | ID: mdl-22151889

ABSTRACT

BACKGROUND: FTY720 (Fingolimod) is a novel immunosuppressive drug investigated in clinical trials for organ transplantation and multiple sclerosis. It acts as a functional sphingosine-1-phosphate (S1P) receptor antagonist, thereby inhibiting the egress of lymphocytes from secondary lymphoid organs. As S1P is able to prevent IL-1beta induced cartilage degradation, we examined the direct impact of FTY720 on cytokine induced cartilage destruction. METHODS: Bovine chondrocytes were treated with the bioactive phosphorylated form of FTY720 (FTY720-P) in combination with IL-1beta or TNF-alpha. Expression of MMP-1,-3.-13, iNOS and ADAMTS-4,-5 and COX-2 was evaluated using quantitative real-time PCR and western blot. Glycosaminoglycan depletion from cartilage explants was determined using a 1,9-dimethylene blue assay and safranin O staining. RESULTS: FTY720-P significantly reduced IL-1beta and TNF-alpha induced expression of iNOS. In contrast FTY720-P increased MMP-3 and ADAMTS-5 mRNA expression. Furthermore depletion of glycosaminoglycan from cartilage explants by IL-1beta and TNF-alpha was significantly enhanced by FTY720-P in an MMP-3 dependent manner. CONCLUSIONS: Our results suggest that FTY720 may enhance cartilage degradation in pro-inflammatory environment.


Subject(s)
Cartilage, Articular/drug effects , Chondrocytes/drug effects , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/metabolism , Immunosuppressive Agents/toxicity , Propylene Glycols/toxicity , Sphingosine/analogs & derivatives , Animals , Cartilage, Articular/enzymology , Cartilage, Articular/metabolism , Cattle , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Fingolimod Hydrochloride , Interleukin-1beta/physiology , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Sphingosine/toxicity , Tumor Necrosis Factor-alpha/physiology
17.
N Engl J Med ; 356(23): 2349-60, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17554116

ABSTRACT

BACKGROUND: Amyloid A (AA) amyloidosis is a complication of chronic inflammatory conditions that develops when proteolytic fragments of serum amyloid A protein (SAA) are deposited in tissues as amyloid fibrils. Amyloid deposition in the kidney causes progressive deterioration in renal function. Eprodisate is a member of a new class of compounds designed to interfere with interactions between amyloidogenic proteins and glycosaminoglycans and thereby inhibit polymerization of amyloid fibrils and deposition of the fibrils in tissues. METHODS: We performed a multicenter, randomized, double-blind, placebo-controlled trial to evaluate the efficacy and safety of eprodisate in patients with AA amyloidosis and kidney involvement. We randomly assigned 183 patients from 27 centers to receive eprodisate or placebo for 24 months. The primary composite end point was an assessment of renal function or death. Disease was classified as worsened if any one of the following occurred: doubling of the serum creatinine level, reduction in creatinine clearance by 50% or more, progression to end-stage renal disease, or death. RESULTS: At 24 months, disease was worsened in 24 of 89 patients who received eprodisate (27%) and 38 of 94 patients given placebo (40%, P=0.06); the hazard ratio for worsening disease with eprodisate treatment was 0.58 (95% confidence interval, 0.37 to 0.93; P=0.02). The mean rates of decline in creatinine clearance were 10.9 and 15.6 ml per minute per 1.73 m(2) of body-surface area per year in the eprodisate and the placebo groups, respectively (P=0.02). The drug had no significant effect on progression to end-stage renal disease (hazard ratio, 0.54; P=0.20) or risk of death (hazard ratio, 0.95; P=0.94). The incidence of adverse events was similar in the two groups. CONCLUSIONS: Eprodisate slows the decline of renal function in AA amyloidosis. (ClinicalTrials.gov number, NCT00035334.)


Subject(s)
Amyloidosis/drug therapy , Glycosaminoglycans/antagonists & inhibitors , Kidney Diseases/drug therapy , Propane/analogs & derivatives , Sulfonic Acids/therapeutic use , Amyloidosis/etiology , Amyloidosis/mortality , Arthritis, Rheumatoid/complications , Creatinine/blood , Disease Progression , Double-Blind Method , Familial Mediterranean Fever/complications , Female , Humans , Kaplan-Meier Estimate , Kidney Diseases/etiology , Kidney Diseases/mortality , Kidney Failure, Chronic/prevention & control , Male , Middle Aged , Propane/adverse effects , Propane/therapeutic use , Proportional Hazards Models , Proteinuria , Serum Amyloid A Protein/drug effects , Sulfonic Acids/adverse effects
18.
Mol Genet Metab ; 101(2-3): 208-13, 2010.
Article in English | MEDLINE | ID: mdl-20650670

ABSTRACT

MPS IIIA is a lysosomal storage disorder caused by mutations in the sulphamidase gene, resulting in the accumulation of heparan sulphate glycosaminoglycans (HS GAGs). Symptoms predominantly manifest in the CNS and there is no current therapy that effectively addresses neuropathology in MPS IIIA patients. Recent studies in MPS IIIA mice have shown that rhodamine B substrate deprivation therapy (SDT) (also termed substrate reduction therapy/SRT) inhibits GAG biosynthesis and, improves both somatic and CNS disease pathology. Acute overexposure to high doses of rhodamine B results in liver toxicity and is detrimental to reproductive ability. However, the long-term effects of decreasing GAG synthesis, at the low dose sufficient to alter neurological function are unknown. A trans-generational study was therefore initiated to evaluate the continuous exposure of rhodamine B treatment in MPS IIIA mice over 4 generations, including treatment during pregnancy. No alterations in litter size, liver histology or liver function were observed. Overall, there are no long-term issues with the administration of rhodamine B at the low dose tested and no adverse effects were noted during pregnancy in mice.


Subject(s)
Glycosaminoglycans/antagonists & inhibitors , Liver/drug effects , Mucopolysaccharidosis III/physiopathology , Rhodamines/therapeutic use , Animals , Disease Models, Animal , Female , Glycosaminoglycans/biosynthesis , Litter Size/drug effects , Liver/pathology , Liver/physiology , Mice , Mucopolysaccharidosis III/genetics , Pregnancy
19.
Bioorg Med Chem Lett ; 20(24): 7269-73, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21074423

ABSTRACT

Various 4-deoxy-4-fluoro-xylosides were prepared using click chemistry for evaluating their potential utility as inhibitors of glycosaminoglycan biosynthesis. 2,3-Di-O-benzoyl-4-deoxy-4-fluoro-ß-D-xylopyranosylazide, obtained from L-arabinopyranose by six steps, was treated with a wide variety of azide-reactive triple bond-containing hydrophobic agents in the presence of Cu(2+) salt/ascorbic acid, a step known as click chemistry. After click chemistry, benzoylated derivatives were deprotected under Zemplén conditions to obtain 4-deoxy-4-fluoro-xyloside derivatives. A mixture of α:ß-isomers of twelve derivatives were then separated on a reverse phase C18 column using HPLC and the resulting twenty four 4-deoxy-4-fluoro-xylosides were evaluated for their ability to inhibit glycosaminoglycan biosynthesis in endothelial cells. We identified two xyloside derivatives that selectively inhibit heparan sulfate and chondroitin sulfate/derman sulfate biosynthesis without affecting cell viability. These novel derivatives can potentially be used to define the biological actions of proteoglycans in model organisms and also as therapeutic agents to combat various human diseases in which glycosaminoglycans participate.


Subject(s)
Glycosaminoglycans/biosynthesis , Glycosides/chemistry , Animals , Azides/chemistry , Catalysis , Cattle , Chondroitin Sulfates/antagonists & inhibitors , Chondroitin Sulfates/biosynthesis , Click Chemistry , Copper/chemistry , Endothelial Cells/cytology , Glycosaminoglycans/antagonists & inhibitors , Glycosides/chemical synthesis , Glycosides/pharmacology , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/biosynthesis , Isomerism
20.
Science ; 175(4024): 898-900, 1972 Feb 25.
Article in English | MEDLINE | ID: mdl-4257647

ABSTRACT

Factors that induce cell aggregation are released by several types of chick embryo and mammalian cell cultures. These aggregation factors are also present in some serums. The factors in each of the preparations tested were inactivated by treatment with bovine testicular hyaluronidase. Conversely, hyaluronic acid promoted aggregation of only those cells that were aggregated by media containing the factors. These factors appear to be acid mucopolysaccharides, with hyaluronic acid being a major component.


Subject(s)
Cell Aggregation/drug effects , Cells, Cultured/analysis , Glycosaminoglycans/pharmacology , Animals , Cell Line , Chick Embryo , Chromatography , Cricetinae , Glycosaminoglycans/antagonists & inhibitors , Glycosaminoglycans/isolation & purification , Humans , Hyaluronic Acid/analysis , Hyaluronic Acid/pharmacology , Hyaluronoglucosaminidase/pharmacology , Mice , Neoplasms, Experimental , Rats
SELECTION OF CITATIONS
SEARCH DETAIL