Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 457
Filter
1.
Chemistry ; 30(31): e202400883, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38556469

ABSTRACT

We report on the syntheses of NeuAc and NeuGc-containing glycosides via the use of double carbonyl-protected N-acetyl sialyl donors. The 7-O,9-O-carbonyl protection of an N-acyl-5-N,4-O-carbonyl-protected sialyl donor markedly increased the α-selectivity during glycosylation, particularly when glycosylating the C-8 hydroxyl group of sialic acids. The N-acyl carbamates were selectively opened with ethanethiol under basic conditions to provide N-acyl amines. It is noteworthy that N-glycolyl carbamate was more reactive to nucleophiles by comparison with the N-acetyl carbamate due to the electron-withdrawing oxygen in the N-acyl group and however, allowed selective opening of the carbamates without the loss of N-glycolyl groups. To demonstrate the utility of the approach, we began by synthesizing α(2,3) and α(2,6) sialyl galactosides. Glycosylation of the hydroxy groups of galactosides at the C-6 position with the NeuAc and NeuGc donors provided the corresponding sialyl galactoses in good yields with excellent α-selectivity. However, glycosylation of the 2,3-diol galactosyl acceptor selectively provided Siaα(2,2)Gal. Next, we prepared a series of α(2,8) disialosides composed of NeuAc and NeuGc. Glycosylation of NeuGc and NeuAc acceptors at the C-8 hydroxyl group with NeuGc and NeuAc sialyl donors provided the corresponding α(2,8) disialosides, and no significant differences were detected in the reactivities of these acceptors.


Subject(s)
Sialic Acids , Glycosylation , Sialic Acids/chemistry , Sialic Acids/chemical synthesis , Carbamates/chemistry , Carbamates/chemical synthesis , Glycosides/chemistry , Glycosides/chemical synthesis , Galactosides/chemistry , Galactosides/chemical synthesis , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/chemical synthesis
2.
Biol Reprod ; 109(6): 799-811, 2023 12 11.
Article in English | MEDLINE | ID: mdl-37672213

ABSTRACT

Galectins are a phylogenetically conserved family of soluble ß-galactoside binding proteins. There are 16 different of galectins, each with a specific function determined by its distinct distribution and spatial structure. Galectin-13, galectin-14, and galectin-16 are distinct from other galectin members in that they are primarily found in placental tissue. These galectins, also referred to as placental galectins, play critical roles in regulating pregnancy-associated processes, such as placenta formation and maternal immune tolerance to the embedded embryo. The unique structural characteristics and the inability to bind lactose of placental galectins have recently received significant attention. This review primarily examines the novel structural features of placental galectins, which distinguish them from the classic galectins. Furthermore, it explores the correlation between these structural features and the loss of ß-galactoside binding ability. In addition, the newly discovered functions of placental galectins in recent years are also summarized in our review. A detailed understanding of the roles of placental galectins may contribute to the discovery of new mechanisms causing numerous pregnancy diseases and enable the development of new diagnostic and therapeutic strategies for the treatment of these diseases, ultimately benefiting the health of mothers and offspring.


Subject(s)
Galectins , Placenta , Female , Pregnancy , Humans , Placenta/metabolism , Galectins/chemistry , Galectins/metabolism , Galactosides/chemistry , Galactosides/metabolism
3.
J Org Chem ; 88(13): 9313-9320, 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37269328

ABSTRACT

Design and synthesis of orthogonally protected monosaccharide building blocks are crucial for the preparation of well-defined oligosaccharides in a stereo- and regiocontrolled manner. Selective introduction of protecting groups to partially protected monosaccharides is nontrivial due to the often unpredictable electronic, steric, and conformational effects of the substituents. Abolished reactivity toward a commonly used Lewis base-catalyzed acylation of O-2 was observed in conformationally restricted 4,6-O-benzylidene-3-O-Nap galactoside. Investigation of analogous systems, crystallographic characterization, and quantum chemical calculations highlighted the overlooked conformational and steric considerations, the combination of which produces a unique passivity of the 2-OH nucleophile. Evaluating the role of electrophile counterion and auxiliary base in the acylation of the sterically crowded and conformationally restricted galactoside system revealed an alternative Brønsted base-driven reaction pathway via nucleophilic activation. Insights gained from this model system were utilized to access the target galactoside intermediate within the envisioned synthetic route. The acylation strategy described herein can be implemented in future syntheses of key monomeric building blocks with unique protecting group hierarchies.


Subject(s)
Galactosides , Galactosides/chemistry , Indicators and Reagents , Acylation
4.
Angew Chem Int Ed Engl ; 62(7): e202215535, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36398566

ABSTRACT

Bacterial adhesion, biofilm formation and host cell invasion of the ESKAPE pathogen Pseudomonas aeruginosa require the tetravalent lectins LecA and LecB, which are therefore drug targets to fight these infections. Recently, we have reported highly potent divalent galactosides as specific LecA inhibitors. However, they suffered from very low solubility and an intrinsic chemical instability due to two acylhydrazone motifs, which precluded further biological evaluation. Here, we isosterically substituted the acylhydrazones and systematically varied linker identity and length between the two galactosides necessary for LecA binding. The optimized divalent LecA ligands showed improved stability and were up to 1000-fold more soluble. Importantly, these properties now enabled their biological characterization. The lead compound L2 potently inhibited LecA binding to lung epithelial cells, restored wound closure in a scratch assay and reduced the invasiveness of P. aeruginosa into host cells.


Subject(s)
Adhesins, Bacterial , Pseudomonas aeruginosa , Humans , Adhesins, Bacterial/chemistry , Pseudomonas aeruginosa/metabolism , Virulence Factors/metabolism , Galactosides/chemistry , Galactosides/metabolism , Galactosides/pharmacology , Bacterial Adhesion
5.
Fish Shellfish Immunol ; 131: 1264-1274, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36400370

ABSTRACT

Galectins are an evolutionarily ancient family of lectins characterized by their affinity for ß-galactosides and a conserved binding site in the carbohydrate recognition domain (CRD). These lectins are involved in multiple physiological functions, including the recognition of glycans on the surface of viruses and bacteria. This feature supports their role in innate immune responses in marine mollusks. Here, we identified and characterized a galectin, from the mollusk Haliotis rufescens (named HrGal), with four CRDs that belong to the tandem-repeat type. HrGal was purified by affinity chromatography in a galactose-agarose resin and exhibited a molecular mass of 64.11 kDa determined by MALDI-TOF mass spectrometry. The identity of HrGal was verified by sequencing, confirming that it is a 555 amino acid protein with a mass of 63.86 kDa. This protein corresponds to a galectin reported in GenBank with accession number AHX26603. HrGal is stable in the presence of urea, reducing agents, and ions such as Cu2+ and Zn2+. The recombinant galectin (rHrGal) was purified from inclusion bodies in the presence of these ions. A theoretical model obtained with the AlphaFold server exhibits four non-identical CRDs, with a ß sandwich folding and the representative motifs for binding ß-galactosides. This allows us to classify HrGal within the tandem repeat galectin family. On the basis of a phylogenetic analysis, we found that the mollusk sequences form a monophyletic group of tetradomain galectins unrelated to vertebrate galectins. HrGal showed specificity for galactosides and glucosides but only the sulfated sugars heparin and ι-carrageenan inhibited its hemagglutinating activity with a minimum inhibitory concentration of 4 mM and 6.25 X 10-5% respectively. The position of the sulfate groups seemed crucial for binding, both by carrageenans and heparin.


Subject(s)
Galectins , Gastropoda , Animals , Galectins/chemistry , Phylogeny , Sulfates , Galactosides/chemistry , Gastropoda/genetics , Gastropoda/metabolism , Polysaccharides , Mollusca/genetics , Heparin
6.
J Biol Chem ; 295(52): 18426-18435, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33127644

ABSTRACT

α-Linked galactose is a common carbohydrate motif in nature that is processed by a variety of glycoside hydrolases from different families. Terminal Galα1-3Gal motifs are found as a defining feature of different blood group and tissue antigens, as well as the building block of the marine algal galactan λ-carrageenan. The blood group B antigen and linear α-Gal epitope can be processed by glycoside hydrolases in family GH110, whereas the presence of genes encoding GH110 enzymes in polysaccharide utilization loci from marine bacteria suggests a role in processing λ-carrageenan. However, the structure-function relationships underpinning the α-1,3-galactosidase activity within family GH110 remain unknown. Here we focus on a GH110 enzyme (PdGH110B) from the carrageenolytic marine bacterium Pseudoalteromonas distincta U2A. We showed that the enzyme was active on Galα1-3Gal but not the blood group B antigen. X-ray crystal structures in complex with galactose and unhydrolyzed Galα1-3Gal revealed the parallel ß-helix fold of the enzyme and the structural basis of its inverting catalytic mechanism. Moreover, an examination of the active site reveals likely adaptations that allow accommodation of fucose in blood group B active GH110 enzymes or, in the case of PdGH110, accommodation of the sulfate groups found on λ-carrageenan. Overall, this work provides insight into the first member of a predominantly marine clade of GH110 enzymes while also illuminating the structural basis of α-1,3-galactoside processing by the family as a whole.


Subject(s)
Blood Group Antigens/metabolism , Carrageenan/metabolism , Galactosides/metabolism , Glycoside Hydrolases/chemistry , Pseudoalteromonas/enzymology , Blood Group Antigens/chemistry , Carrageenan/chemistry , Catalytic Domain , Crystallography, X-Ray , Galactosides/chemistry , Glycoside Hydrolases/classification , Glycoside Hydrolases/metabolism , Hydrolysis , Models, Molecular , Phylogeny , Protein Conformation , Substrate Specificity
7.
Glycobiology ; 31(11): 1490-1499, 2021 12 18.
Article in English | MEDLINE | ID: mdl-34255029

ABSTRACT

Pseudomonas aeruginosa is a widespread opportunistic pathogen that is capable of colonizing various human tissues and is resistant to many antibiotics. LecA is a galactose binding tetrameric lectin involved in adhesion, infection and biofilm formation. This study reports on the binding characteristics of mono- and divalent (chelating) ligands to LecA using different techniques. These techniques include affinity capillary electrophoresis, bio-layer interferometry, native mass spectrometry and a thermal shift assay. Aspects of focus include: affinity, selectivity, binding kinetics and residence time. The affinity of a divalent ligand was determined to be in the low-nanomolar range for all of the used techniques and with a ligand residence time of approximately 7 h, while no strong binding was seen to related lectin tetramers. Each of the used techniques provides a unique and complementary insight into the chelation based binding mode of the divalent ligand to the LecA tetramer.


Subject(s)
Galactosides/chemistry , Lectins/chemistry , Pseudomonas aeruginosa/chemistry , Temperature , Binding Sites , Electrophoresis, Capillary , Interferometry , Ligands , Mass Spectrometry
8.
Bioorg Med Chem ; 44: 116309, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34293617

ABSTRACT

Galectins, soluble lectins widely expressed intra- and extracellularly in different cell types, play major roles in deciphering the cellular glycocode. Galectin-1 (Gal-1), a prototype member of this family, presents a carbohydrate recognition domain (CRD) with specific affinity for ß-galactosides such as N-acetyllactosamine (ß-d-Galp-(1 â†’ 4)-d-GlcpNAc), and mediate numerous physiological and pathological processes. In this work, Gal-1 binding affinity for ß-(1 â†’ 6) galactosides, including ß-d-Galp-(1 â†’ 6)-ß-d-GlcpNAc-(1 â†’ 4)-d-GlcpNAc was evaluated, and their performance was compared to that of ß-(1 â†’ 4) and ß-(1 â†’ 3) galactosides. To this end, the trisaccharide ß-d-Galp-(1 â†’ 6)-ß-d-GlcpNAc-(1 â†’ 4)-d-GlcpNAc was enzymatically synthesized, purified and structurally characterized. To evaluate the affinity of Gal-1 for the galactosides, competitive solid phase assays (SPA) and isothermal titration calorimetry (ITC) studies were carried out. The experimental dissociation constants and binding energies obtained were compared to those calculated by molecular docking. These analyses evidenced the critical role of the glycosidic linkage between the terminal galactopyranoside residue and the adjacent monosaccharide, as galactosides bearing ß-(1 â†’ 6) glycosidic linkages showed dissociation constants six- and seven-fold higher than those involving ß-(1 â†’ 4) and ß-(1 â†’ 3) linkages, respectively. Moreover, docking experiments revealed the presence of hydrogen bond interactions between the N-acetyl group of the glucosaminopyranose moiety of the evaluated galactosides and specific amino acid residues of Gal-1, relevant for galectin-glycan affinity. Noticeably, the binding free energies (ΔGbindcalc) derived from the molecular docking were in good agreement with experimental values determined by ITC measurements (ΔGbindexp), evidencing a good correlation between theoretical and experimental approaches, which validates the in silico simulations and constitutes an important tool for the rational design of future optimized ligands.


Subject(s)
Galactosides/chemistry , Galectin 1/chemistry , Sugars/chemistry , Acetylation , Carbohydrate Conformation , Humans , Molecular Docking Simulation
9.
Anal Bioanal Chem ; 413(9): 2361-2388, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33606064

ABSTRACT

ß-Galactosidase (ß-Gal) is a widely used enzyme as a reporter gene in the field of molecular biology which hydrolyzes the ß-galactosides into monosaccharides. ß-Gal is an essential enzyme in humans and its deficiency or its overexpression results in several rare diseases. Cellular senescence is probably one of the most relevant physiological disorders that involve ß-Gal enzyme. In this review, we assess the progress made to date in the design of molecular-based probes for the detection of ß-Gal both in vitro and in vivo. Most of the reported molecular probes for the detection of ß-Gal consist of a galactopyranoside residue attached to a signalling unit through glycosidic bonds. The ß-Gal-induced hydrolysis of the glycosidic bonds released the signalling unit with remarkable changes in color and/or emission. Additional examples based on other approaches are also described. The wide applicability of these probes for the rapid and in situ detection of de-regulation ß-Gal-related diseases has boosted the research in this fertile field.


Subject(s)
Fluorescent Dyes/chemistry , Galactose/analogs & derivatives , beta-Galactosidase/analysis , Animals , Cellular Senescence , Colorimetry/methods , Enzyme Assays/methods , Galactosides/chemistry , Humans , Hydrolysis , Molecular Probes/chemistry
10.
Mediators Inflamm ; 2021: 7372893, 2021.
Article in English | MEDLINE | ID: mdl-33551679

ABSTRACT

BACKGROUND: Our previous research showed the antioxidant activity of anthocyanins extracted from Aronia melanocarpa of black chokeberry in vitro. Ischemia acute kidney injury is a significant risk in developing progressive and deterioration of renal function leading to clinic chronic kidney disease. There were many attempts to protect the kidney against this progression of renal damage. Current study was designed to examine the effect of pretreatment with three anthocyanins named cyanidin-3-arabinoside, cyanidin-3-glucodise, and cyaniding-3-galactoside against acute ischemia-reperfusion injury in mouse kidney. METHODS: Acute renal injury model was initiated by 30 min clamping bilateral renal pedicle and followed by 24-hour reperfusion in C57Bl/6J mice. Four groups of mice were orally pretreated in 50 mg/g/12 h for two weeks with cyanidin-3-arabinoside, cyanidin-3-glucodise, and cyaniding-3-galactoside and anthocyanins (three-cyanidin mixture), respectively, sham-control group and the renal injury-untreated groups only with saline. RESULTS: The model resulted in renal dysfunction with high serum creatinine, blood urea nitrogen, and changes in proinflammatory cytokines (TNF-ɑ, IL-1ß, IL-6, and MCP-1), renal oxidative stress (SOD, GSH, and CAT), lipid peroxidation (TBARS and MDA), and apoptosis (caspase-9). Pretreatment of two weeks resulted in different extent amelioration of renal dysfunction and tubular damage and suppression of proinflammatory cytokines, oxidative stress, lipid peroxidation, and apoptosis, thus suggesting that cyanidins are potentially effective in acute renal ischemia by the decrease of inflammation, oxidative stress, and lipid peroxidation, as well as apoptosis. CONCLUSION: the current study provided the first attempt to investigate the role of anthocyanins purified from Aronia melanocarpa berry in amelioration of acute renal failure via antioxidant and cytoprotective effects.


Subject(s)
Anthocyanins/metabolism , Kidney Failure, Chronic/metabolism , Kidney/drug effects , Photinia/metabolism , Reperfusion Injury , Animals , Anthocyanins/chemistry , Antioxidants/chemistry , Apoptosis , Arabinonucleosides/chemistry , Body Weight , Caspase 9/metabolism , Fruit , Galactosides/chemistry , Inflammation , Kidney/metabolism , Lipid Peroxidation , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Reperfusion , Risk
11.
Proc Natl Acad Sci U S A ; 115(50): 12716-12721, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30478058

ABSTRACT

The lactose permease of Escherichia coli (LacY) utilizes an alternating access symport mechanism with multiple conformational intermediates, but only inward (cytoplasmic)- or outward (periplasmic)-open structures have been characterized by X-ray crystallography. It is demonstrated here with sugar-binding studies that cross-linking paired-Cys replacements across the closed cytoplasmic cavity stabilize an occluded conformer with an inaccessible sugar-binding site. In addition, a nanobody (Nb) that stabilizes a periplasmic-open conformer with an easily accessible sugar-binding site in WT LacY fails to cause the cytoplasmic cross-linked mutants to become accessible to galactoside, showing that the periplasmic cavity is closed. These results are consistent with tight association of the periplasmic ends in two pairs of helices containing clusters of small residues in the packing interface between N- and C-terminal six-helix bundles of the symporter. However, after reduction of the disulfide bond, the Nb markedly increases the rate of galactoside binding, indicating unrestricted access to the Nb epitope and the galactoside-binding site from the periplasm. The findings indicate that the cross-linked cytoplasmic double-Cys mutants resemble an occluded apo-intermediate in the transport cycle.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Monosaccharide Transport Proteins/chemistry , Symporters/chemistry , Binding Sites , Crystallography, X-Ray/methods , Cytoplasm/metabolism , Escherichia coli/metabolism , Galactosides/chemistry , Galactosides/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Periplasm/metabolism , Symporters/metabolism
12.
Proc Natl Acad Sci U S A ; 115(16): 4146-4151, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29602806

ABSTRACT

Binding kinetics of α-galactopyranoside homologs with fluorescent aglycones of different sizes and shapes were determined with the lactose permease (LacY) of Escherichia coli by FRET from Trp151 in the binding site of LacY to the fluorophores. Fast binding was observed with LacY stabilized in an outward-open conformation (kon = 4-20 µM-1·s-1), indicating unobstructed access to the binding site even for ligands that are much larger than lactose. Dissociation rate constants (koff) increase with the size of the aglycone so that Kd values also increase but remain in the micromolar range for each homolog. Phe27 (helix I) forms an apparent constriction in the pathway for sugar by protruding into the periplasmic cavity. However, replacement of Phe27 with a bulkier Trp does not create an obstacle in the pathway even for large ligands, since binding kinetics remain unchanged. High accessibility of the binding site is also observed in a LacY/nanobody complex with partially blocked periplasmic opening. Remarkably, E. coli expressing WT LacY catalyzes transport of α- or ß-galactopyranosides with oversized aglycones such as bodipy or Aldol518, which may require an extra space within the occluded intermediate. The results confirm that LacY specificity is strictly directed toward the galactopyranoside ring and also clearly indicate that the opening on the periplasmic side is sufficiently wide to accommodate the large galactoside derivatives tested here. We conclude that the actual pathway for the substrate entering from the periplasmic side is wider than the pore diameter calculated in the periplasmic-open X-ray structures.


Subject(s)
Escherichia coli Proteins/metabolism , Galactosides/metabolism , Monosaccharide Transport Proteins/metabolism , Symporters/metabolism , Binding Sites , Biological Transport, Active , Crystallography, X-Ray , Escherichia coli Proteins/chemistry , Fluorescent Dyes , Galactose/chemistry , Galactose/metabolism , Galactosides/chemistry , Kinetics , Ligands , Models, Molecular , Molecular Structure , Monosaccharide Transport Proteins/chemistry , Periplasm/metabolism , Protein Binding , Protein Conformation , Structure-Activity Relationship , Symporters/chemistry
13.
Proc Natl Acad Sci U S A ; 115(12): E2819-E2828, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507247

ABSTRACT

Treatment of bacterial infections is becoming a serious clinical challenge due to the global dissemination of multidrug antibiotic resistance, necessitating the search for alternative treatments to disarm the virulence mechanisms underlying these infections. Uropathogenic Escherichia coli (UPEC) employs multiple chaperone-usher pathway pili tipped with adhesins with diverse receptor specificities to colonize various host tissues and habitats. For example, UPEC F9 pili specifically bind galactose or N-acetylgalactosamine epitopes on the kidney and inflamed bladder. Using X-ray structure-guided methods, virtual screening, and multiplex ELISA arrays, we rationally designed aryl galactosides and N-acetylgalactosaminosides that inhibit the F9 pilus adhesin FmlH. The lead compound, 29ß-NAc, is a biphenyl N-acetyl-ß-galactosaminoside with a Ki of ∼90 nM, representing a major advancement in potency relative to the characteristically weak nature of most carbohydrate-lectin interactions. 29ß-NAc binds tightly to FmlH by engaging the residues Y46 through edge-to-face π-stacking with its A-phenyl ring, R142 in a salt-bridge interaction with its carboxylate group, and K132 through water-mediated hydrogen bonding with its N-acetyl group. Administration of 29ß-NAc in a mouse urinary tract infection (UTI) model significantly reduced bladder and kidney bacterial burdens, and coadministration of 29ß-NAc and mannoside 4Z269, which targets the type 1 pilus adhesin FimH, resulted in greater elimination of bacteria from the urinary tract than either compound alone. Moreover, FmlH specifically binds healthy human kidney tissue in a 29ß-NAc-inhibitable manner, suggesting a key role for F9 pili in human kidney colonization. Thus, these glycoside antagonists of FmlH represent a rational antivirulence strategy for UPEC-mediated UTI treatment.


Subject(s)
Adhesins, Escherichia coli/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Urinary Tract Infections/microbiology , Adhesins, Escherichia coli/metabolism , Animals , Crystallography, X-Ray , Drug Evaluation, Preclinical/methods , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Female , Galactosides/chemical synthesis , Galactosides/chemistry , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/microbiology , Ligands , Mice, Inbred C3H , Molecular Docking Simulation , Molecular Mimicry , Urinary Tract Infections/drug therapy , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/pathogenicity
14.
Mikrochim Acta ; 189(1): 4, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34855041

ABSTRACT

Synthetic biology approaches for rewiring of bacterial constructs to express particular intracellular factors upon induction with the target analyte are emerging as sensing paradigms for applications in environmental and in vivo monitoring. To aid in the design and optimization of bacterial constructs for sensing analytes, there is a need for lysis-free intracellular detection modalities that monitor the signal level and kinetics of expressed factors within different modified bacteria in a multiplexed manner, without requiring cumbersome surface immobilization. Herein, an electrochemical detection system on nanoporous gold that is electrofabricated with a biomaterial redox capacitor is presented for quantifying ß-galactosidase expressed inside modified Escherichia coli constructs upon induction with dopamine. This nanostructure-mediated redox amplification approach on a microfluidic platform allows for multiplexed assessment of the expressed intracellular factors from different bacterial constructs suspended in distinct microchannels, with no need for cell lysis or immobilization. Since redox mediators present over the entire depth of the microchannel can interact with the electrode and with the E. coli construct in each channel, the platform exhibits high sensitivity and enables multiplexing. We envision its application in assessing synthetic biology-based approaches for comparing specificity, sensitivity, and signal response time upon induction with target analytes of interest.


Subject(s)
Catechols/chemistry , Chitosan/chemistry , Electrochemical Techniques/methods , Escherichia coli Proteins/analysis , Nanopores , beta-Galactosidase/analysis , Dopamine/pharmacology , Escherichia coli/enzymology , Escherichia coli Proteins/metabolism , Galactosides/chemistry , Galactosides/metabolism , Gold/chemistry , Limit of Detection , Microfluidic Analytical Techniques , Oxidation-Reduction , Ruthenium/chemistry , Trans-Activators/metabolism , beta-Galactosidase/metabolism
15.
Int J Mol Sci ; 22(9)2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33925857

ABSTRACT

Building-up and breaking-down of carbohydrates are processes common to all forms of life. Glycoside hydrolases are a broad class of enzymes that play a central role in the cleavage of glycosidic bonds, which is fundamental to carbohydrate degradation. The large majority of substrates are five- and six-membered ring glycosides. Our interest in seven-membered ring septanose sugars has inspired the development of a way to search for septanoside hydrolase activity. Described here is a strategy for the discovery of septanoside hydrolases that uses synthetic indolyl septanosides as chromogenic substrates. Access to these tool compounds was enabled by a route where septanosyl halides act as glycosyl donors for the synthesis of the indolyl septanosides. The screening strategy leverages the known dimerization of 3-hydroxy-indoles to make colored dyes, as occurs when the ß-galactosidase substrate X-Gal is hydrolyzed. Because screens in bacterial cells would enable searches in organisms that utilize heptoses or from metagenomics libraries, we also demonstrate that septanosides are capable of entering E. coli cells through the use of a BODIPY-labeled septanoside. The modularity of the indolyl septanoside synthesis should allow the screening of a variety of substrates that mimic natural structures via this general approach.


Subject(s)
Escherichia coli/metabolism , Glycosides/biosynthesis , Hydrolases/metabolism , Carbohydrate Metabolism , Chromogenic Compounds/chemistry , Escherichia coli/chemistry , Galactosides/biosynthesis , Galactosides/chemistry , Glycoside Hydrolases/metabolism , Glycosides/chemistry , Hydrolysis , Indoles/chemistry
16.
J Biol Chem ; 294(31): 11701-11711, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31186348

ABSTRACT

Bifidobacteria are exposed to substantial amounts of dietary ß-galactosides. Distinctive preferences for growth on different ß-galactosides are observed within Bifidobacterium members, but the basis of these preferences remains unclear. We previously described the first ß-(1,6)/(1,3)-galactosidase from Bifidobacterium animalis subsp. lactis Bl-04. This enzyme is relatively promiscuous, exhibiting only 5-fold higher efficiency on the preferred ß-(1,6)-galactobiose than the ß-(1,4) isomer. Here, we characterize the solute-binding protein (Bal6GBP) that governs the specificity of the ABC transporter encoded by the same ß-galactoside utilization locus. We observed that although Bal6GBP recognizes both ß-(1,6)- and ß-(1,4)-galactobiose, Bal6GBP has a 1630-fold higher selectivity for the former, reflected in dramatic differences in growth, with several hours lag on less preferred ß-(1,4)- and ß-(1,3)-galactobiose. Experiments performed in the presence of varying proportions of ß-(1,4)/ß-(1,6)-galactobioses indicated that the preferred substrate was preferentially depleted from the culture supernatant. This established that the poor growth on the nonpreferred ß-(1,4) was due to inefficient uptake. We solved the structure of Bal6GBP in complex with ß-(1,6)-galactobiose at 1.39 Å resolution, revealing the structural basis of this strict selectivity. Moreover, we observed a close evolutionary relationship with the human milk disaccharide lacto-N-biose-binding protein from Bifidobacterium longum, indicating that the recognition of the nonreducing galactosyl is essentially conserved, whereas the adjacent position is diversified to fit different glycosidic linkages and monosaccharide residues. These findings indicate that oligosaccharide uptake has a pivotal role in governing selectivity for distinct growth substrates and have uncovered evolutionary trajectories that shape the diversification of sugar uptake proteins within Bifidobacterium.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Bifidobacterium animalis/growth & development , Galactosidases/metabolism , Galactosides/metabolism , ATP-Binding Cassette Transporters/chemistry , Amino Acid Sequence , Bacterial Proteins/chemistry , Bifidobacterium animalis/enzymology , Bifidobacterium animalis/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Evolution, Molecular , Galactosidases/chemistry , Galactosides/chemistry , Kinetics , Molecular Dynamics Simulation , Protein Binding , Substrate Specificity
17.
Org Biomol Chem ; 18(26): 5017-5033, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32573638

ABSTRACT

We developed an indirect synthetic method for α-l-fucosides. Based on the fact that l-fucose is 6-deoxy-l-galactose, our strategy consists of the stereoselective construction of α-l-galactoside and its conversion to α-l-fucoside via C6-deoxygenation. The formation of α-l-galactoside is strongly directed using 4,6-O-di-tert-butylsilylene(DTBS)-protected l-galactosyl donors. The DTBS-directed α-l-galactosylation showed broad substrate applicability along with excellent coupling yield and α-selectivity. In the C6-deoxygenation of α-l-galactosides, the Barton-McCombie reaction facilitated the conversion to l-fucosides with good yield. To demonstrate the applicability of our method, we synthesized naturally occurring α-l-fucosides.


Subject(s)
Fucose/chemical synthesis , Galactosides/chemistry , Oxygen/chemistry , Carbohydrate Conformation , Fucose/chemistry , Glycosylation , Stereoisomerism
18.
Org Biomol Chem ; 18(7): 1462-1475, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32025679

ABSTRACT

Selective glycosylation of the C-6 fluorinated galactofuranosyl acceptor 2 was studied with four galactofuranosyl donors. It was highlighted that this electron-withdrawing atom strongly impacted the behavior of the acceptor, thus leading to unprecedented glycosylation pathways. Competition between expected glycosylation of 2, ring expansion of this acceptor and furanosylation, and intermolecular aglycon transfer was observed. Further investigation of the fluorinated synthetic compounds showed that the presence of fluorine atom contributed to increase the inhibition of the growth of Leishmania tarentolae, a non-pathogenic strain of Leishmania.


Subject(s)
Antiprotozoal Agents/pharmacology , Furans/pharmacology , Galactosides/pharmacology , Leishmania/drug effects , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Carbohydrate Conformation , Furans/chemical synthesis , Furans/chemistry , Galactosides/chemical synthesis , Galactosides/chemistry , Glycosylation , Parasitic Sensitivity Tests , Stereoisomerism
19.
Biosci Biotechnol Biochem ; 84(4): 797-799, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31790630

ABSTRACT

Insect gall structures have many characteristic forms and colors, which are distinguishable from host plants. In this study, we identified an anthocyanin from red color insect galls and revealed that the anthocyanin biosynthesis of plants was induced by the gall extracts. The galling insects presumably regulate the anthocyanin biosynthesis of host plants to protect their larvae from environmental stresses.


Subject(s)
Anthocyanins/chemistry , Ceratopogonidae/physiology , Fagus/parasitology , Galactosides/chemistry , Host-Parasite Interactions , Animals , Anthocyanins/biosynthesis , Ceratopogonidae/growth & development , Fagus/metabolism , Larva/physiology
20.
Biotechnol Lett ; 42(11): 2379-2387, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32654007

ABSTRACT

OBJECTIVE: To synthesize octyl ß-D-glucopyranoside (OG) and decyl ß-D-glucopyranoside (DG) in three non-aqueous reaction systems, namely organic solvents, ionic liquids and co-solvent mixtures, via reverse hydrolysis reactions catalyzed by the N189F dalcochinase mutant. RESULTS: The highest yield of OG (67 mol%) was obtained in the reaction containing 0.5 M glucose, 3 unit ml-1 enzyme in 20% (v/v) octanol and 70% (v/v) [BMIm][PF6] at 30 °C. On the other hand, the highest yield of DG (64 mol%) was obtained in the reaction containing 0.5 M glucose, 3 unit ml-1 enzyme in 20% (v/v) decanol, 20% (v/v) acetone and 50% (v/v) [BMIm][PF6] at 30 °C. The identities of OG and DG products were confirmed by HRMS and NMR. CONCLUSION: This is the first report of enzymatic synthesis of OG and DG via reverse hydrolysis reactions in ionic liquids and co-solvent mixtures. The N189F dalcochinase mutant and the non-aqueous reaction systems described here show great potential for future commercial production of long-chain alkyl glucosides.


Subject(s)
Galactosides/chemistry , Solvents/chemistry , beta-Glucosidase/metabolism , Hydrolysis , Ionic Liquids/chemistry , Protein Engineering
SELECTION OF CITATIONS
SEARCH DETAIL