Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 14(10): 1411-1418, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37849547

ABSTRACT

Heparan sulfate-mimicking glycopolymers, composed of glucosamine (GlcN)-glucuronic acid (GlcA) repeating units, bind to the receptor-binding subunit (S1) and spike glycoprotein (S) domains of the SARS-CoV-2 spike protein in a length- and sulfation pattern-dependent fashion. A glycopolymer composed of 12 repeating GlcNS6S-GlcA units exhibits a much higher affinity to the S1 protein (IC50 = 13 ± 1.1 nM) compared with the receptor-binding domain (RBD). This glycopolymer does not interfere in angiotensin-converting enzyme 2 binding of the RBD. Although this compound binds strongly to the S1/membrane-fusion subunit (S2) junction (KD = 29.7 ± 4.18 nM), it does not shield the S1/S2 site from cleavage by furin-a behavior contrary to natural heparin. This glycopolymer lacks iduronic acid, which accounts for 70% of heparin. Further, this compound, unlike natural heparin, is well defined in both sulfation pattern and length, which results in fewer off-target interactions with heparin-binding proteins. The results highlight the potential of using polymeric heparan sulfate (HS) mimetics for the therapeutic agent development.

2.
ACS Chem Biol ; 17(6): 1387-1400, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35658404

ABSTRACT

Diabetes is a chronic disease in which the levels of blood glucose are too high because the body does not effectively produce insulin to meet its needs or is resistant to insulin. ß Cells in human pancreatic islets produce insulin, which signals glucogen production by the liver and causes muscles and fat to uptake glucose. Progressive loss of insulin-producing ß cells is the main cause of both type 1 and type 2 diabetes. Heparan sulfate (HS) is a ubiquitous polysaccharide found at the cell surface and in the extracellular matrix (ECM) of a variety of tissues. HS binds to and assembles proteins in ECM, thus playing important roles in the integrity of ECM (particularly basement membrane), barrier function, and ECM-cell interactions. Islet HS is highly expressed by the pancreatic ß cells and critical for the survival of ß cells. Heparanase is an endoglycosidase and cleaves islet HS in the pancreas, resulting in ß-cell death and oxidative stress. Heparanase could also accelerate ß-cell death by promoting cytokine release from ECM and secretion by activated inflammatory and endothelial cells. We demonstrate that HS-mimicking glycopolymer, a potent heparanase inhibitor, improves the survival of cultured mouse pancreatic ß cells and protects HS contents under the challenge of heparanase in human pancreatic islets. Moreover, this HS-mimicking glycopolymer reduces the expression levels of cytokines (IL8, IL1ß, and TNFα) and the gene encoding Toll-like Receptor 2 (TLR2) in human pancreatic islets.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Biomimetic Materials/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Glucuronidase , Heparitin Sulfate/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice
3.
J Med Chem ; 63(8): 4227-4255, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32216347

ABSTRACT

Heparanase cleaves polymeric heparan sulfate (HS) molecules into smaller oligosaccharides, allowing for release of angiogenic growth factors promoting tumor development and autoreactive immune cells to reach the insulin-producing ß cells. Interaction of heparanase with HS chains is regulated by specific substrate sulfation sequences. We have synthesized 11 trisaccharides that are highly tunable in structure and sulfation pattern, allowing us to determine how heparanase recognizes HS substrate and selects a favorable cleavage site. Our study shows that (1) N-SO3- at +1 subsite and 6-O-SO3- at -2 subsite of trisaccharides are critical for heparanase recognition, (2) addition of 2-O-SO3- at the -1 subsite and of 3-O-SO3- to GlcN unit is not advantageous, and (3) the anomeric configuration (α or ß) at the reducing end is crucial in controlling heparanase activity. Our study also illustrates that the α-trisaccharide having N- and 6-O-SO3- at -2 and +1 subsites inhibited heparanase and was resistant toward hydrolysis.


Subject(s)
Enzyme Activation/physiology , Glucuronidase/metabolism , Glycosides/metabolism , Heparitin Sulfate/metabolism , Oligosaccharides/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glycosides/chemical synthesis , Heparin/pharmacology , Heparitin Sulfate/antagonists & inhibitors , Humans , Mice , Molecular Docking Simulation/methods , Oligosaccharides/chemical synthesis
4.
ACS Catal ; 10(11): 5990-6001, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-34168901

ABSTRACT

Copper-catalyzed cross-coupling reactions have become one of the most powerful methods for generating carbon-heteroatom bonds, an important framework of many organic molecules. However, copper-catalyzed C(sp3)-O cross-coupling of alkyl halides with alkyl alcohols remains elusive because of the sluggish nature of oxidative addition to copper. To address this challenge, we have developed a catalytic copper system, which overcomes the copper oxidative addition barrier with the aid of visible light and effectively facilitates the cross-couplings of glycosyl bromides with aliphatic alcohols to afford C(sp3)-O bonds with high levels of diastereoselectivity. Importantly, this catalytic system leads to a mild and efficient method for stereoselective construction of α-1,2-cis glycosides, which are of paramount importance, but challenging. In general, stereochemical outcomes in α-1,2-cis glycosidic C-O bond-forming processes are unpredictable and dependent on the steric and electronic nature of protecting groups bound to carbohydrate coupling partners. Currently, the most reliable approaches rely on the use of a chiral auxiliary or hydrogen-bond directing group at the C2- and C4-position of carbohydrate electrophiles to control α-1,2-cis selectivity. In our approach, earth-abundant copper not only acts as a photocatalyst and a bond-forming catalyst, but also enforces the stereocontrolled formation of anomeric C-O bonds. This cross-coupling protocol enables highly diastereoselective access to a wide variety of α-1,2-cis-glycosides and biologically relevant α-glycan oligosaccharides. Our work provides a foundation for developing new methods for the stereoselective construction of natural and unnatural anomeric carbon(sp3)-heteroatom bonds.

5.
ACS Appl Mater Interfaces ; 11(1): 244-254, 2019 Jan 09.
Article in English | MEDLINE | ID: mdl-30543095

ABSTRACT

Heparanase, the heparan sulfate polysaccharide degrading endoglycosidase enzyme, has been correlated with tumor angiogenesis and metastasis and therefore has become a potential target for anticancer drug development. In this systematic study, the sulfation pattern of the pendant disaccharide moiety on synthetic glycopolymers was synthetically manipulated to achieve optimal heparanase inhibition. Upon evaluation, a glycopolymer with 12 repeating units was determined to be the most potent inhibitor of heparanase (IC50 = 0.10 ± 0.36 nM). This glycopolymer was further examined for cross-bioactivity using a solution-based competitive biolayer interferometry assay with other HS-binding proteins (growth factors, P-selectin, and platelet factor 4), which are responsible for mediating angiogenic activity, cell metastasis, and antibody-induced thrombocytopenia. The synthetic glycopolymer has low affinity for these HS-binding proteins in comparison to natural heparin. In addition, the glycopolymer possessed no proliferative properties toward human umbilical endothelial cells (HUVECs) and a potent antimetastatic effect against 4T1 mammary carcinoma cells. Thus, our study not only establishes a specific inhibitor of heparanase with high affinity but also illustrates the high effectiveness of this multivalent heparanase inhibitor in inhibiting experimental metastasis in vivo.


Subject(s)
Enzyme Inhibitors/pharmacology , Glucuronidase , Mammary Neoplasms, Experimental , Neoplasm Proteins , Neovascularization, Pathologic , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Female , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Heparin/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , P-Selectin/metabolism
6.
Chem Commun (Camb) ; 53(65): 9163-9166, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28766595

ABSTRACT

Heparanase is an enzyme which cleaves heparan sulfate (HS) polysaccharides of the extracellular matrix. It is a regulator of tumor behavior, plays a key role in kidney related diseases and autoimmune diabetes. We report herein the use of computational studies to extract the natural HS-heparanase interactions as a template for the design of HS mimicking glycopolymers. Upon evaluation, a glycopolymer with 12 repeating units was determined to be the most potent inhibitor and to have tight-binding characteristics. This glycopolymer also lacks anticoagulant activity.


Subject(s)
Biomimetic Materials/chemistry , Enzyme Inhibitors/chemistry , Glucuronidase/antagonists & inhibitors , Polysaccharides/chemistry , Biomimetic Materials/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Factor Xa Inhibitors/chemical synthesis , Factor Xa Inhibitors/chemistry , Heparitin Sulfate/chemistry , Molecular Docking Simulation , Polysaccharides/chemical synthesis , Prothrombin/antagonists & inhibitors
7.
Biomacromolecules ; 18(10): 3387-3399, 2017 Oct 09.
Article in English | MEDLINE | ID: mdl-28846389

ABSTRACT

We report herein the first-time exploration of the attachment of well-defined saccharide units onto a synthetic polymer backbone for the inhibition of a glycosidase. More specifically, glycopolymers endowed with heparan sulfate (HS) disaccharides were established to inhibit the glycosidase, heparanase, with an IC50 value in the low nanomolar range (1.05 ± 0.02 nm), a thousand-fold amplification over its monovalent counterpart. The monomeric moieties of these glycopolymers were designed in silico to manipulate the well-established glycotope of heparanase into an inhitope. Studies concluded that (1) the glycopolymers are hydrolytic stable toward heparanase, (2) longer polymer length provides greater inhibition, and (3) increased local saccharide density (monoantennary vs diantennary) is negligible due to hindered active site of heparanase. Furthermore, HS oligosaccharide and polysaccharide controls illustrate the enhanced potency of a multivalent scaffold. Overall, the results on these studies of the multivalent presentation of saccharides on bottlebrush polymers serve as the platform for the design of potent glycosidase inhibitors and have potential to be applied to other HS-degrading proteins.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Heparitin Sulfate/chemistry , Binding Sites , Enzyme Inhibitors/pharmacology , Glucuronidase/antagonists & inhibitors , Polymerization , Protein Binding
8.
Biomacromolecules ; 16(12): 4013-4021, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26580410

ABSTRACT

Preparations of the highly ordered monoantennary, homofunctional diantennary, and heterofunctional diantennary neoglycopolymers of α-d-mannose and ß-d-glucose residues were achieved via ring-opening metathesis polymerization. Isothermal titration calorimetry measurements of these synthetic neoglycopolymers with Concanavalin A (Con A), revealed that heterofunctional diantennary architectures bearing both α-mannose and nonbinding ß-glucose units, poly(Man-Glc), binds to Con A (Ka = 16.1 × 10(6) M(-1)) comparably to homofunctional diantennary neoglycopolymer (Ka = 30 × 10(6) M(-1)) bearing only α-mannose unit, poly(Man-Man). In addition, poly(Man-Glc) neoglycopolymer shows a nearly 5-fold increasing in binding affinity compared to monoantennary neoglycopolymer, poly(Man). Although the exact mechanism for the high binding affinity of poly(Man-Glc) to Con A is unclear, we hypothesize that the α-mannose bound to Con A might facilitate interaction of ß-glucose with the extended binding site of Con A due to the close proximity of ß-glucose to α-mannose residues in the designed polymerizable scaffold.


Subject(s)
Concanavalin A/chemistry , Glucose/chemistry , Mannose/chemistry , Binding Sites , Calorimetry/methods , Carbohydrate Conformation , Kinetics , Polymerization
9.
Bioconjug Chem ; 24(6): 907-14, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23742724

ABSTRACT

The glycan of specific proteins can dictate the response of cells to stimuli, and thus their phenotype. We describe a chemical strategy to modify the cellular glycoform of T cells, which resulted in a modified cellular response. Our data indicate that chemical modification of the phosphatase CD45 is responsible for the observed differences in response to receptor cross-linking. By increasing the content of galactose epitopes in the glycocalyx of a lymphoma cell line, we were able to increase the response of the cell to lectin stimulation through the glycoprotein receptor, CD45. The method described here exploits metabolic labeling of a cell to reprogram the cellular response to external stimuli though changes in the number of lectin binding sites on the cell surface.


Subject(s)
Epitopes, T-Lymphocyte/metabolism , Leukocyte Common Antigens/metabolism , Polysaccharides/metabolism , Cells, Cultured , Epitopes, T-Lymphocyte/chemistry , Humans , Jurkat Cells , Leukocyte Common Antigens/chemistry , Molecular Structure , Phenotype , Polysaccharides/chemistry
10.
Carbohydr Res ; 345(18): 2641-7, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20971453

ABSTRACT

The detection of carbohydrate-protein interactions is often performed using techniques that require surface immobilization of the lectin or the glycan. A commonly used assay for lectin binding is surface plasmon resonance (SPR). We describe an implementation of the Staudinger ligation as a method to immobilize carbohydrate epitopes to a biosensor surface. This was accomplished by first introducing an azide functionality to a carboxymethyldextran surface, followed by reaction with a phosphane-modified carbohydrate ligand. The chemistry employed is extremely mild and was easily adapted to a commercial biosensor system. Using this approach, we investigated the binding of jacalin and wheat germ agglutinin (WGA) to galactose, lactose, and N-acetyl-lactosamine. We observed that WGA binding shows evidence of multivalent interaction with the surface. Additionally, we found that jacalin binding was influenced by the presence of a flexible and hydrophobic galactosyl aglycone.


Subject(s)
Epitopes/chemistry , Surface Plasmon Resonance/methods , Lectins/chemistry , Molecular Structure , Plant Lectins/chemistry , Wheat Germ Agglutinins/chemistry
11.
Bioconjug Chem ; 21(10): 1842-9, 2010 Oct 20.
Article in English | MEDLINE | ID: mdl-20845977

ABSTRACT

Generation of defined glycoconjugates is necessary for the study of glycoprotein function, as well as the development of therapeutics. The biosynthesis of glycoproteins produces multiple glycoforms, proteins which differ only in the structure of the attached glycan. This inherent heterogeneity complicates the study of isolated glycans and, in particular, could obscure the role of individual glycan epitopes in biological function. We present a general strategy based on the Staudinger ligation to introduce specific glycan epitopes onto azide-containing proteins. The use of a phosphane-based Staudinger reagent allows for extremely mild reaction conditions which can be applied to aqueous proteins or cells. We demonstrate that multiple carbohydrate epitopes can be incorporated onto a protein backbone, and that the resulting glycans are competent for recognition by lectins. We propose that this general strategy will allow for testing the role of specific glycan epitopes in cellular and biochemical assays and increasing the stability of protein conjugates.


Subject(s)
Amino Sugars/chemistry , Azides/chemistry , Glycoproteins/chemistry , Amines/chemistry , Amino Sugars/chemical synthesis , Disaccharides/chemistry , Epitopes/chemistry , Epitopes/metabolism , Glycoproteins/metabolism , Indicators and Reagents/chemistry , Monosaccharides/chemistry , Phosphines/chemistry , Plant Lectins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...