Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 18(1): 151-165, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36626752

ABSTRACT

Altered cellular metabolism is a hallmark of cancer pathogenesis and progression; for example, a near-universal feature of cancer is increased metabolic flux through the hexosamine biosynthetic pathway (HBP). This pathway produces uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), a potent oncometabolite that drives multiple facets of cancer progression. In this study, we synthesized and evaluated peracetylated hexosamine analogs designed to reduce flux through the HBP. By screening a panel of analogs in pancreatic cancer and glioblastoma multiform (GBM) cells, we identified Ac4Glc2Bz─a benzyl-modified GlcNAc mimetic─as an antiproliferative cancer drug candidate that down-regulated oncogenic metabolites and reduced GBM cell motility at concentrations non-toxic to non-neoplastic cells. More specifically, the growth inhibitory effects of Ac4Glc2Bz were linked to reduced levels of UDP-GlcNAc and concomitant decreases in protein O-GlcNAc modification in both pancreatic cancer and GBM cells. Targeted metabolomics analysis in GBM cells showed that Ac4Glc2Bz disturbed glucose metabolism, amino acid pools, and nucleotide precursor biosynthesis, consistent with reduced proliferation and other anti-oncogenic properties of this analog. Furthermore, Ac4Glc2Bz reduced the invasion, migration, and stemness of GBM cells. Importantly, normal metabolic functions mediated by UDP-GlcNAc were not disrupted in non-neoplastic cells, including maintenance of endogenous levels of O-GlcNAcylation with no global disruption of N-glycan production. Finally, a pilot in vivo study showed that a potential therapeutic window exists where animals tolerated 5- to 10-fold higher levels of Ac4Glc2Bz than projected for in vivo efficacy. Together, these results establish GlcNAc analogs targeting the HBP through salvage mechanisms as a new therapeutic approach to safely normalize an important facet of aberrant glucose metabolism associated with cancer.


Subject(s)
Antineoplastic Agents , Pancreatic Neoplasms , Animals , Biosynthetic Pathways , Hexosamines/metabolism , Antineoplastic Agents/pharmacology , Glucose/metabolism , Uridine Diphosphate/metabolism , Acetylglucosamine/metabolism , Pancreatic Neoplasms
2.
Methods Mol Biol ; 2442: 391-411, 2022.
Article in English | MEDLINE | ID: mdl-35320537

ABSTRACT

Galectin-3 is a chimeric galectin involved in diverse intracellular and extracellular functions. Galectin-3 is synthesized in the cytoplasm and then released extracellularly by a poorly understood non-canonical secretion mechanism. As a result, it can play important roles both inside and outside the cell. One important extracellular role of galectin-3 is in modulating clathrin-independent endocytosis (CIE), a form of cellular internalization that is still not well understood. CIE, unlike clathrin-mediated endocytosis, has neither defined signaling sequences nor cytoplasmic machinery. As a result, extracellular interactions like the galectin-glycan interactions are thought to directly drive changes in CIE. This chapter discusses the methods designed to study the role of galectin-glycan interactions in CIE, which have provided us with insight into the functions of galectin-3 and cell surface glycans during CIE cargo internalization. These methods include media supplementation for metabolic glycoengineering, antibody internalization assays, lectin panels to assay changes in glycan patterns, exogenous galectin-3 supplementation, galectin-3 secretion assays, and in vitro assays to monitor the effect of galectins on CIE.


Subject(s)
Clathrin , Endocytosis , Galectin 3 , Cell Membrane/metabolism , Clathrin/metabolism , Endocytosis/physiology , Galectin 3/metabolism , Methods
3.
J Biol Chem ; 298(3): 101743, 2022 03.
Article in English | MEDLINE | ID: mdl-35183508

ABSTRACT

Endomembrane glycosylation and cytoplasmic O-GlcNAcylation each play essential roles in nutrient sensing, and characteristic changes in glycan patterns have been described in disease states such as diabetes and cancer. These changes in glycosylation have important functional roles and can drive disease progression. However, little is known about the molecular mechanisms underlying how these signals are integrated and transduced into biological effects. Galectins are proteins that bind glycans and that are secreted by a poorly characterized nonclassical secretory mechanism. Once outside the cell, galectins bind to the terminal galactose residues of cell surface glycans and modulate numerous extracellular functions, such as clathrin-independent endocytosis (CIE). Originating in the cytoplasm, galectins are predicted substrates for O-GlcNAc addition and removal; and as we have shown, galectin 3 is a substrate for O-GlcNAc transferase. In this study, we also show that galectin 3 secretion is sensitive to changes in O-GlcNAc levels. We determined using immunoprecipitation and Western blotting that there is a significant difference in O-GlcNAcylation status between cytoplasmic and secreted galectin 3. We observed dramatic alterations in galectin 3 secretion in response to nutrient conditions, which were dependent on dynamic O-GlcNAcylation. Importantly, we showed that these O-GlcNAc-driven alterations in galectin 3 secretion also facilitated changes in CIE. These results indicate that dynamic O-GlcNAcylation of galectin 3 plays a role in modulating its secretion and can tune its function in transducing nutrient-sensing information coded in cell surface glycosylation into biological effects.


Subject(s)
Galectin 3 , Acetylglucosamine/metabolism , Clathrin/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Glycosylation , N-Acetylglucosaminyltransferases/metabolism , Nutrients , Polysaccharides/metabolism , Protein Processing, Post-Translational
4.
Traffic ; 20(4): 295-300, 2019 04.
Article in English | MEDLINE | ID: mdl-30706592

ABSTRACT

In contrast to clathrin-mediated endocytosis (CME) which is well characterized and understood, little is known about the regulation and machinery underlying clathrin-independent endocytosis (CIE). There is also a wide variation in the requirements each individual CIE cargo has for its internalization. Recent studies have shown that CIE is affected by glycosylation and glycan interactions. We briefly review these studies and explore how these studies mesh with one another. We then discuss what this sensitivity to glycan interactions could indicate for the regulation of CIE. We address the spectrum of responses CIE has been shown to have with respect to changes in glycan interactions and attempt to reconcile disparate observations onto a shared conceptual landscape. We focus on the mechanisms by which cells can alter the glycan interactions at the plasma membrane and propose that glycosylation and glycan interactions could provide cells with a tool box with which cells can manipulate CIE. Altered glycosylation is often associated with a number of diseases and we discuss how under different disease settings, glycosylation-based modulation of CIE could play a role in disease progression.


Subject(s)
Endocytosis , Galectins/metabolism , Polysaccharides/metabolism , Protein Processing, Post-Translational , Animals , Glycosylation , Humans , Membrane Proteins/metabolism
5.
Acta Biomater ; 72: 228-238, 2018 05.
Article in English | MEDLINE | ID: mdl-29631048

ABSTRACT

There has been growing interest in the use of particles coated with lipids for applications ranging from drug delivery, gene delivery, and diagnostic imaging to immunoengineering. To date, almost all particles with lipid coatings have been spherical despite emerging evidence that non-spherical shapes can provide important advantages including reduced non-specific elimination and increased target-specific binding. We combine control of core particle geometry with control of particle surface functionality by developing anisotropic, biodegradable ellipsoidal particles with lipid coatings. We demonstrate that these lipid coated ellipsoidal particles maintain advantageous properties of lipid polymer hybrid particles, such as the ability for modular protein conjugation to the particle surface using versatile bioorthogonal ligation reactions. In addition, they exhibit biomimetic membrane fluidity and demonstrate lateral diffusive properties characteristic of natural membrane proteins. These ellipsoidal particles simultaneously provide benefits of non-spherical particles in terms of stability and resistance to non-specific phagocytosis by macrophages as well as enhanced targeted binding. These biomaterials provide a novel and flexible platform for numerous biomedical applications. STATEMENT OF SIGNIFICANCE: The research reported here documents the ability of non-spherical polymeric particles to be coated with lipids to form anisotropic biomimetic particles. In addition, we demonstrate that these lipid-coated biodegradable polymeric particles can be conjugated to a wide variety of biological molecules in a "click-like" fashion. This is of interest due to the multiple types of cellular mimicry enabled by this biomaterial based technology. These features include mimicry of the highly anisotropic shape exhibited by cells, surface presentation of membrane bound protein mimetics, and lateral diffusivity of membrane bound substrates comparable to that of a plasma membrane. This platform is demonstrated to facilitate targeted cell binding while being resistant to non-specific cellular uptake. Such a platform could allow for investigations into how physical parameters of a particle and its surface affect the interface between biomaterials and cells, as well as provide biomimetic technology platforms for drug delivery and cellular engineering.


Subject(s)
Coated Materials, Biocompatible , Lipids , Macrophages/metabolism , Membranes, Artificial , Phagocytosis/drug effects , Proteins , Animals , Anisotropy , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacokinetics , Coated Materials, Biocompatible/pharmacology , Lipids/chemistry , Lipids/pharmacokinetics , Lipids/pharmacology , Macrophages/cytology , Mice , Proteins/chemistry , Proteins/pharmacokinetics , Proteins/pharmacology , RAW 264.7 Cells
6.
J Biol Chem ; 293(19): 7222-7237, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29581232

ABSTRACT

Clathrin-independent endocytosis (CIE) is a form of endocytosis that lacks a defined cytoplasmic machinery. Here, we asked whether glycan interactions, acting from the outside, could be a part of that endocytic machinery. We show that the perturbation of global cellular patterns of protein glycosylation by modulation of metabolic flux affects CIE. Interestingly, these changes in glycosylation had cargo-specific effects. For example, in HeLa cells, GlcNAc treatment, which increases glycan branching, increased major histocompatibility complex class I (MHCI) internalization but inhibited CIE of the glycoprotein CD59 molecule (CD59). The effects of knocking down the expression of galectin 3, a carbohydrate-binding protein and an important player in galectin-glycan interactions, were also cargo-specific and stimulated CD59 uptake. By contrast, inhibition of all galectin-glycan interactions by lactose inhibited CIE of both MHCI and CD59. None of these treatments affected clathrin-mediated endocytosis, implying that glycosylation changes specifically affect CIE. We also found that the galectin lattice tailors membrane fluidity and cell spreading. Furthermore, changes in membrane dynamics mediated by the galectin lattice affected macropinocytosis, an altered form of CIE, in HT1080 cells. Our results suggest that glycans play an important and nuanced role in CIE, with each cargo being affected uniquely by alterations in galectin and glycan profiles and their interactions. We conclude that galectin-driven effects exist on a continuum from stimulatory to inhibitory, with distinct CIE cargo proteins having unique response landscapes and with different cell types starting at different positions on these conceptual landscapes.


Subject(s)
Endocytosis/physiology , Galectin 3/metabolism , Polysaccharides/metabolism , Acetylglucosamine/pharmacology , CD59 Antigens/metabolism , Cell Membrane/drug effects , Cell Movement/physiology , Clathrin/physiology , Culture Media , Galectin 3/genetics , Galectin 3/pharmacology , Gene Knockdown Techniques , Glycosylation , HeLa Cells , Histocompatibility Antigens Class I/metabolism , Humans , Lactose/pharmacology , Membrane Fluidity/physiology , Pinocytosis/physiology , Protein Transport/physiology
7.
Chembiochem ; 18(13): 1204-1215, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28218815

ABSTRACT

This report describes the metabolic glycoengineering (MGE) of intracellular esterase activity in human colon cancer (LS174T) and Chinese hamster ovary (CHO) cells. In silico analysis of carboxylesterases CES1 and CES2 suggested that these enzymes are modified with sialylated N-glycans, which are proposed to stabilize the active multimeric forms of these enzymes. This premise was supported by treating cells with butanolylated ManNAc to increase sialylation, which in turn increased esterase activity. By contrast, hexosamine analogues not targeted to sialic acid biosynthesis (e.g., butanoylated GlcNAc or GalNAc) had minimal impact. Measurement of mRNA and protein confirmed that esterase activity was controlled through glycosylation and not through transcription or translation. Azide-modified ManNAc analogues widely used in MGE also enhanced esterase activity and provided a way to enrich targeted glycoengineered proteins (such as CES2), thereby providing unambiguous evidence that the compounds were converted to sialosides and installed into the glycan structures of esterases as intended. Overall, this study provides a pioneering example of the modulation of intracellular enzyme activity through MGE, which expands the value of this technology from its current status as a labeling strategy and modulator of cell surface biological events.


Subject(s)
Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Epithelial Cells/enzymology , Metabolic Engineering/methods , Protein Processing, Post-Translational , Sialic Acids/metabolism , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Acetylgalactosamine/pharmacology , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Acetylglucosamine/pharmacology , Animals , Binding Sites , Butyric Acid/chemistry , CHO Cells , Carboxylesterase/chemistry , Carboxylesterase/genetics , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/genetics , Cell Line, Tumor , Cricetulus , Epithelial Cells/cytology , Epithelial Cells/drug effects , Glycosylation , Hexosamines/chemistry , Hexosamines/metabolism , Hexosamines/pharmacology , Humans , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Sialic Acids/chemistry
8.
Biomaterials ; 116: 158-173, 2017 02.
Article in English | MEDLINE | ID: mdl-27926828

ABSTRACT

Abnormal cell surface display of sialic acids - a family of unusual 9-carbon sugars - is widely recognized as distinguishing feature of many types of cancer. Sialoglycans, however, typically cannot be identified with sufficiently high reproducibility and sensitivity to serve as clinically accepted biomarkers and similarly, almost all efforts to exploit cancer-specific differences in sialylation signatures for therapy remain in early stage development. In this report we provide an overview of important facets of glycosylation that contribute to cancer in general with a focus on breast cancer as an example of malignant disease characterized by aberrant sialylation. We then describe how cancer cells experience nutrient deprivation during oncogenesis and discuss how the resulting metabolic reprogramming, which endows breast cancer cells with the ability to obtain nutrients during scarcity, constitutes an "Achilles' heel" that we believe can be exploited by metabolic glycoengineering (MGE) strategies to develop new diagnostic methods and therapeutic approaches. In particular, we hypothesize that adaptations made by breast cancer cells that allow them to efficiently scavenge sialic acid during times of nutrient deprivation renders them vulnerable to MGE, which refers to the use of exogenously-supplied, non-natural monosaccharide analogues to modulate targeted aspects of glycosylation in living cells and animals. In specific, once non-natural sialosides are incorporated into the cancer "sialome" they can be exploited as epitopes for immunotherapy or as chemical tags for targeted delivery of imaging or therapeutic agents selectively to tumors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Metabolic Engineering/methods , N-Acetylneuraminic Acid/biosynthesis , N-Acetylneuraminic Acid/genetics , Theranostic Nanomedicine/methods , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
9.
Oncotarget ; 7(41): 66491-66511, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27613843

ABSTRACT

In prior work we reported that advanced stage, drug-resistant pancreatic cancer cells (the SW1990 line) can be sensitized to the EGFR-targeting tyrosine kinase inhibitors (TKIs) erlotinib and gefitinib by treatment with 1,3,4-O-Bu3ManNAc (Bioorg. Med. Chem. Lett. (2015) 25(6):1223-7). Here we provide mechanistic insights into how this compound inhibits EGFR activity and provides synergy with TKI drugs. First, we showed that the sialylation of the EGFR receptor was at most only modestly enhanced (by ~20 to 30%) compared to overall ~2-fold increase in cell surface levels of this sugar. Second, flux-driven sialylation did not alter EGFR dimerization as has been reported for cancer cell lines that experience increased sialylation due to spontaneous mutations. Instead, we present evidence that 1,3,4-O-Bu3ManNAc treatment weakens the galectin lattice, increases the internalization of EGFR, and shifts endosomal trafficking towards non-clathrin mediated (NCM) endocytosis. Finally, by evaluating downstream targets of EGFR signaling, we linked synergy between 1,3,4-O-Bu3ManNAc and existing TKI drugs to a shift from clathrin-coated endocytosis (which allows EGFR signaling to continue after internalization) towards NCM endocytosis, which targets internalized moieties for degradation and thereby rapidly diminishes signaling.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , ErbB Receptors/metabolism , Pancreatic Neoplasms/metabolism , Protein Transport/drug effects , Cell Line, Tumor , Endocytosis/drug effects , Humans , Protein Kinase Inhibitors/pharmacology
10.
Data Brief ; 5: 481-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26629491

ABSTRACT

This report provides data that are specifically related to the differential sialylation of nutrient deprived breast cancer cells to sialic acid supplementation in support of the research article entitled, "Nutrient-deprived cancer cells preferentially use sialic acid to maintain cell surface glycosylation" [1]. Particularly, breast cancer cells, when supplemented with sialic acid under nutrient deprivation, display sialylated glycans at the cell surface, but non-malignant mammary cells show sialylated glycans intracellularly. The impact of sialic acid supplementation under nutrient deprivation was demonstrated by measuring levels of expression and sialylation of two markers, EGFR1 and MUC1. This Data in Brief article complements the main manuscript by providing detailed instructions and representative results for cell-level imaging and Western blot analyses of changes in sialylation during nutrient deprivation and sialic acid supplementation. These methods can be readily generalized for the study of many types of glycosylation and various glycoprotein markers through the appropriate selection of fluorescently-labeled lectins.

11.
Biomaterials ; 70: 23-36, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26295436

ABSTRACT

Cancer is characterized by abnormal energy metabolism shaped by nutrient deprivation that malignant cells experience during various stages of tumor development. This study investigated the response of nutrient-deprived cancer cells and their non-malignant counterparts to sialic acid supplementation and found that cells utilize negligible amounts of this sugar for energy. Instead cells use sialic acid to maintain cell surface glycosylation through complementary mechanisms. First, levels of key metabolites (e.g., UDP-GlcNAc and CMP-Neu5Ac) required for glycan biosynthesis are maintained or enhanced upon Neu5Ac supplementation. In concert, sialyltransferase expression increased at both the mRNA and protein levels, which facilitated increased sialylation in biochemical assays that measure sialyltransferase activity as well as at the whole cell level. In the course of these experiments, several important differences emerged that differentiated the cancer cells from their normal counterparts including resistant to sialic acid-mediated energy depletion, consistently more robust sialic acid-mediated glycan display, and distinctive cell surface vs. internal vesicle display of newly-produced sialoglycans. Finally, the impact of sialic acid supplementation on specific markers implicated in cancer progression was demonstrated by measuring levels of expression and sialylation of EGFR1 and MUC1 as well as the corresponding function of sialic acid-supplemented cells in migration assays. These findings both provide fundamental insight into the biological basis of sialic acid supplementation of nutrient-deprived cancer cells and open the door to the development of diagnostic and prognostic tools.


Subject(s)
Cell Membrane/metabolism , N-Acetylneuraminic Acid/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Adenosine Triphosphate/metabolism , Blotting, Western , Cell Line, Tumor , Cell Membrane/chemistry , Cell Movement , Cell Survival , ErbB Receptors/metabolism , Glycoconjugates/metabolism , Glycosylation , Humans , Lectins/metabolism , Monosaccharides/metabolism , Mucin-1/metabolism , Nucleotides/metabolism , Real-Time Polymerase Chain Reaction , Sialic Acids/metabolism , Sialyltransferases/metabolism
12.
J Funct Biomater ; 6(2): 454-85, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26096148

ABSTRACT

Membranes constitute the interface between the basic unit of life-a single cell-and the outside environment and thus in many ways comprise the ultimate "functional biomaterial". To perform the many and often conflicting functions required in this role, for example to partition intracellular contents from the outside environment while maintaining rapid intake of nutrients and efflux of waste products, biological membranes have evolved tremendous complexity and versatility. This article describes how membranes, mainly in the context of living cells, are increasingly being manipulated for practical purposes with drug discovery, biofuels, and biosensors providing specific, illustrative examples. Attention is also given to biology-inspired, but completely synthetic, membrane-based technologies that are being enabled by emerging methods such as bio-3D printers. The diverse set of applications covered in this article are intended to illustrate how these versatile technologies-as they rapidly mature-hold tremendous promise to benefit human health in numerous ways ranging from the development of new medicines to sensitive and cost-effective environmental monitoring for pathogens and pollutants to replacing hydrocarbon-based fossil fuels.

13.
Glycoconj J ; 32(7): 425-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25931032

ABSTRACT

Metabolic glycoengineering is a specialization of metabolic engineering that focuses on using small molecule metabolites to manipulate biosynthetic pathways responsible for oligosaccharide and glycoconjugate production. As outlined in this article, this technique has blossomed in mammalian systems over the past three decades but has made only modest progress in prokaryotes. Nevertheless, a sufficient foundation now exists to support several important applications of metabolic glycoengineering in bacteria based on methods to preferentially direct metabolic intermediates into pathways involved in lipopolysaccharide, peptidoglycan, teichoic acid, or capsule polysaccharide production. An overview of current applications and future prospects for this technology are provided in this report.


Subject(s)
Carbohydrate Metabolism/genetics , Glycoproteins/genetics , Metabolic Engineering , Recombinant Proteins/metabolism , Animals , Glycoconjugates/chemistry , Glycoconjugates/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Glycosylation , Lipopolysaccharides/chemistry , Lipopolysaccharides/genetics , Lipopolysaccharides/metabolism , Oligosaccharides/chemical synthesis , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
14.
Bioorg Med Chem Lett ; 25(6): 1223-7, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25690786

ABSTRACT

Metastatic human pancreatic cancer cells (the SW1990 line) that are resistant to the EGFR-targeting tyrosine kinase inhibitor drugs (TKI) erlotinib and gefitinib were treated with 1,3,4-O-Bu3ManNAc, a 'metabolic glycoengineering' drug candidate that increased sialylation by ∼2-fold. Consistent with genetic methods previously used to increase EGFR sialylation, this small molecule reduced EGF binding, EGFR transphosphorylation, and downstream STAT activation. Significantly, co-treatment with both the sugar pharmacophore and the existing TKI drugs resulted in strong synergy, in essence re-sensitizing the SW1990 cells to these drugs. Finally, 1,3,4-O-Bu3ManNAz, which is the azido-modified counterpart to 1,3,4-O-Bu3ManNAc, provided a similar benefit thereby establishing a broad-based foundation to extend a 'metabolic glycoengineering' approach to clinical applications.


Subject(s)
Erlotinib Hydrochloride/chemistry , Metabolic Engineering , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Gefitinib , Glycosylation , Humans , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , STAT Transcription Factors/metabolism
15.
Bioorg Med Chem Lett ; 22(22): 6929-33, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23041156

ABSTRACT

This report provides a synopsis of the esterase processing of short chain fatty acid (SCFA)-derivatized hexosamine analogs used in metabolic glycoengineering by demonstrating that the extracellular hydrolysis of these compounds is comparatively slow (e.g., with a t(1/2) of ∼4 h to several days) in normal cell culture as well as in high serum concentrations intended to mimic in vivo conditions. Structure-activity relationship (SAR) analysis of common sugar analogs revealed that O-acetylated and N-azido ManNAc derivatives were more refractory against extracellular inactivation by FBS than their butanoylated counterparts consistent with in silico docking simulations of Ac(4)ManNAc and Bu(4)ManNAc to human carboxylesterase 1 (hCE1). By contrast, all analogs tested supported increased intracellular sialic acid production within 2h establishing that esterase processing once the analogs are taken up by cells is not rate limiting.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Drug Carriers/chemistry , Fatty Acids, Volatile/chemistry , Hexosamines/chemistry , Binding Sites , Cell Line, Tumor , Hexosamines/metabolism , Humans , Hydrolysis , Molecular Docking Simulation , N-Acetylneuraminic Acid/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
16.
Ann Biomed Eng ; 40(4): 806-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22037949

ABSTRACT

Metabolic oligosaccharide engineering is an emerging technology wherein non-natural monosaccharide analogs are exogenously supplied to living cells and are biosynthetically incorporated into cell surface glycans. A recently reported application of this methodology employs fluorinated analogs of ManNAc, GlcNAc, and GalNAc to modulate selectin-mediated adhesion associated with leukocyte extravasation and cancer cell metastasis. This monograph outlines possible mechanisms underlying the altered adhesion observed in analog-treated cells; these range from the most straightforward explanation (e.g., structural changes to the selectin ligands ablate interaction with their receptors) to the alternative mechanism where the analogs inhibit or otherwise perturb ligand production to more indirect mechanisms (e.g., changes to the biophysical properties of the selectin binding partner, the nanoenviroment of the binding partners, or the entire cell surface).


Subject(s)
Leukocytes/metabolism , Metabolic Engineering/methods , Oligosaccharides/biosynthesis , Selectins/metabolism , Transendothelial and Transepithelial Migration , Animals , Cell Adhesion , Humans , Leukocytes/cytology
SELECTION OF CITATIONS
SEARCH DETAIL