Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters

Publication year range
1.
Microb Cell Fact ; 22(1): 241, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-38012629

ABSTRACT

BACKGROUND: In whole-cell bio-catalysis, the biosystems engineering paradigm shifts from the global reconfiguration of cellular metabolism as in fermentation to a more focused, and more easily modularized, optimization of comparably short cascade reactions. Human milk oligosaccharides (HMO) constitute an important field for the synthetic application of cascade bio-catalysis in resting or non-living cells. Here, we analyzed the central catalytic module for synthesis of HMO-type sialo-oligosaccharides, comprised of CMP-sialic acid synthetase (CSS) and sialyltransferase (SiaT), with the specific aim of coordinated enzyme co-expression in E. coli for reaction flux optimization in whole cell conversions producing 3'-sialyllactose (3SL). RESULTS: Difference in enzyme specific activity (CSS from Neisseria meningitidis: 36 U/mg; α2,3-SiaT from Pasteurella dagmatis: 5.7 U/mg) was compensated by differential protein co-expression from tailored plasmid constructs, giving balance between the individual activities at a high level of both (α2,3-SiaT: 9.4 × 102 U/g cell dry mass; CSS: 3.4 × 102 U/g cell dry mass). Finally, plasmid selection was guided by kinetic modeling of the coupled CSS-SiaT reactions in combination with comprehensive analytical tracking of the multistep conversion (lactose, N-acetyl neuraminic acid (Neu5Ac), cytidine 5'-triphosphate; each up to 100 mM). The half-life of SiaT in permeabilized cells (≤ 4 h) determined the efficiency of 3SL production at 37 °C. Reaction at 25 °C gave 3SL (40 ± 4 g/L) in ∼ 70% yield within 3 h, reaching a cell dry mass-specific productivity of ∼ 3 g/(g h) and avoiding intermediary CMP-Neu5Ac accumulation. CONCLUSIONS: Collectively, balanced co-expression of CSS and SiaT yields an efficient (high-flux) sialylation module to support flexible development of E. coli whole-cell catalysts for sialo-oligosaccharide production.


Subject(s)
Escherichia coli , N-Acylneuraminate Cytidylyltransferase , Humans , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Escherichia coli/metabolism , Oligosaccharides/metabolism , Bioengineering , Sialyltransferases/genetics , Sialyltransferases/metabolism , Catalysis
2.
Biochem Biophys Res Commun ; 617(Pt 1): 16-21, 2022 08 20.
Article in English | MEDLINE | ID: mdl-35667241

ABSTRACT

The CMP-sialic acid synthetase (CSS) activates free sialic acid (Sia) to CMP-Sia using CTP, and is prerequisite for the sialylation of cell surface glycoconjugates. The vertebrate CSS consists of two domains, a catalytic N-domain and a non-catalytic C-domain. Although the C-domain is not required for the CSS enzyme to synthesize CMP-Sia, its involvement in the catalytic activity remains unknown. First, the real-time monitoring of CSS-catalyzed reaction was performed by 31P NMR using the rainbow trout CSS (rtCSS). While a rtCSS lacking the C-domain (rtCSS-N) similarly activated both deaminoneuraminic acid (Kdn) and N-acetylneuraminic acid (Neu5Ac), the full-length rtCSS (rtCSS-FL) did not activate Kdn as efficiently as Neu5Ac. These results suggest that the C-domain of rtCSS affects the enzymatic activity, when Kdn was used as a substrate. Second, the enzymatic activity of rtCSS-FL and rtCSS-N was measured under various concentrations of CMP-Kdn. Inhibition by CMP-Kdn was observed only for rtCSS-FL, but not for rtCSS-N, suggesting that the inhibition was C-domain-dependent. Third, the inhibitory effect of CMP-Kdn was also investigated using the mouse CSS (mCSS). However, no inhibition was observed with mCSS even at high concentrations of CMP-Kdn. Taken together, the data demonstrated that the C-domain is involved in the CMP-Kdn-dependent inhibition of rtCSS, which is a novel regulation of the Sia metabolism in rainbow trout.


Subject(s)
N-Acylneuraminate Cytidylyltransferase , Oncorhynchus mykiss , Animals , Cytidine Monophosphate/analogs & derivatives , Mice , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Neuraminic Acids , Sialic Acids/metabolism
3.
J Virol ; 95(2)2020 12 22.
Article in English | MEDLINE | ID: mdl-33087464

ABSTRACT

Engagement of cell surface receptors by viruses is a critical determinant of viral tropism and disease. The reovirus attachment protein σ1 binds sialylated glycans and proteinaceous receptors to mediate infection, but the specific requirements for different cell types are not entirely known. To identify host factors required for reovirus-induced cell death, we conducted a CRISPR-knockout screen targeting over 20,000 genes in murine microglial BV2 cells. Candidate genes required for reovirus to cause cell death were highly enriched for sialic acid synthesis and transport. Two of the top candidates identified, CMP N-acetylneuraminic acid synthetase (Cmas) and solute carrier family 35 member A1 (Slc35a1), promote sialic acid expression on the cell surface. Two reovirus strains that differ in the capacity to bind sialic acid, T3SA+ and T3SA-, were used to evaluate Cmas and Slc35a1 as potential host genes required for reovirus infection. Following CRISPR-Cas9 disruption of either gene, cell surface expression of sialic acid was diminished. These results correlated with decreased binding of strain T3SA+, which is capable of engaging sialic acid. Disruption of either gene did not alter the low-level binding of T3SA-, which does not engage sialic acid. Furthermore, infectivity of T3SA+ was diminished to levels similar to those of T3SA- in cells lacking Cmas and Slc35a1 by CRISPR ablation. However, exogenous expression of Cmas and Slc35a1 into the respective null cells restored sialic acid expression and T3SA+ binding and infectivity. These results demonstrate that Cmas and Slc35a1, which mediate cell surface expression of sialic acid, are required in murine microglial cells for efficient reovirus binding and infection.IMPORTANCE Attachment factors and receptors are important determinants of dissemination and tropism during reovirus-induced disease. In a CRISPR cell survival screen, we discovered two genes, Cmas and Slc35a1, which encode proteins required for sialic acid expression on the cell surface and mediate reovirus infection of microglial cells. This work elucidates host genes that render microglial cells susceptible to reovirus infection and expands current understanding of the receptors on microglial cells that are engaged by reovirus. Such knowledge may lead to new strategies to selectively target microglial cells for oncolytic applications.


Subject(s)
N-Acylneuraminate Cytidylyltransferase/metabolism , Nucleotide Transport Proteins/metabolism , Reoviridae Infections/virology , Reoviridae/physiology , Animals , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cell Membrane/metabolism , Cell Survival , Mice , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , Nucleotide Transport Proteins/genetics , Receptors, Virus/metabolism , Reoviridae/genetics , Reoviridae/metabolism , Reoviridae Infections/metabolism , Virus Attachment , Virus Replication
4.
Int J Mol Sci ; 22(11)2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34200006

ABSTRACT

Influenza A viruses (IAVs) initiate infection by attaching Hemagglutinin (HA) on the viral envelope to sialic acid (SA) receptors on the cell surface. Importantly, HA of human IAVs has a higher affinity for α-2,6-linked SA receptors, and avian strains prefer α-2,3-linked SA receptors, whereas swine strains have a strong affinity for both SA receptors. Host gene CMAS and ST3GAL4 were found to be essential for IAV attachment and entry. Loss of CMAS and ST3GAL4 hindered the synthesis of sialic acid receptors, which in turn prevented the adsorption of IAV. Further, the knockout of CMAS had an effect on the adsorption of swine, avian and human IAVs. However, ST3GAL4 knockout prevented the adsorption of swine and avian IAV and the impact on avian IAV was more distinct, whereas it had no effect on the adsorption of human IAV. Collectively, our findings demonstrate that knocking out CMAS and ST3GAL4 negatively regulated IAV replication by inhibiting the synthesis of SA receptors, which also provides new insights into the production of gene-edited animals in the future.


Subject(s)
Influenza A virus/physiology , N-Acylneuraminate Cytidylyltransferase/antagonists & inhibitors , Orthomyxoviridae Infections/virology , Receptors, Cell Surface/metabolism , Sialyltransferases/antagonists & inhibitors , Virus Replication , Animals , CRISPR-Cas Systems , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Swine
5.
Biochemistry ; 59(34): 3157-3168, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31583886

ABSTRACT

Cytidine 5'-monophosphate (CMP)-sialic acid synthetase (CSS) is an essential enzyme involved in the biosynthesis of carbohydrates and glycoconjugates containing sialic acids, a class of α-keto acids that are generally terminal key recognition residues by many proteins that play important biological and pathological roles. The CSS from Neisseria meningitidis (NmCSS) has been commonly used with other enzymes such as sialic acid aldolase and/or sialyltransferase in synthesizing a diverse array of compounds containing sialic acid or its naturally occurring and non-natural derivatives. To better understand its catalytic mechanism and substrate promiscuity, four NmCSS crystal structures trapped at various stages of the catalytic cycle with bound substrates, substrate analogues, and products have been obtained and are presented here. These structures suggest a mechanism for an "open" and "closed" conformational transition that occurs as sialic acid binds to the NmCSS/cytidine-5'-triphosphate (CTP) complex. The closed conformation positions critical residues to help facilitate the nucleophilic attack of sialic acid C2-OH to the α-phosphate of CTP, which is also aided by two observed divalent cations. Product formation drives the active site opening, promoting the release of products.


Subject(s)
Biocatalysis , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/enzymology , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Mutation , N-Acylneuraminate Cytidylyltransferase/genetics
6.
Glycobiology ; 27(4): 329-341, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27986833

ABSTRACT

The occurrence and biological importance of sialic acid (Sia) and its metabolic enzymes in insects have been studied using Drosophila melanogaster. The most prominent feature of D. melanogaster CMP-Sia synthetase (DmCSS) is its Golgi-localization, contrasted with nuclear localization of vertebrate CSSs. However, it remains unclear if the Golgi-localization is common to other insect CSSs and why it happens. To answer these questions, Aedes aegypti (mosquito) CSS (AaCSS) and Tribolium castaneum (beetle) CSS (TcCSS) were cloned and characterized for their activity and subcellular localization. Our new findings show: (1) AaCSS and TcCSS share a common overall structure with DmCSS in terms of evolutionarily conserved motifs and the absence of the C-terminal domain typical to vertebrate CSSs; (2) when expressed in mammalian and insect cells, AaCSS and TcCSS showed in vivo and in vitro CSS activities, similar to DmCSS. In contrast, when expressed in bacteria, they lacked CSS activity because the N-terminal hydrophobic region appeared to induce protein aggregation; (3) when expressed in Drosophila S2 cells, AaCSS and TcCSS were predominantly localized in the ER, but not in the Golgi. Surprisingly, DmCSS was mainly secreted into the culture medium, although partially detected in Golgi. Consistent with these results, the N-terminal hydrophobic regions of AaCSS and TcCSS functioned as a signal peptide to render them soluble in the ER, while the N-terminus of DmCSS functioned as a membrane-spanning region of type II transmembrane proteins whose cytosolic KLK sequence functioned as an ER export signal. Accordingly, the differential subcellular localization of insect CSSs are distinctively more diverse than previously recognized.


Subject(s)
N-Acetylneuraminic Acid/genetics , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/genetics , Aedes/enzymology , Amino Acid Motifs/genetics , Animals , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Golgi Apparatus/enzymology , Golgi Apparatus/genetics , Mutation , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Protein Conformation , Tribolium/enzymology
7.
Chembiochem ; 18(13): 1251-1259, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28395125

ABSTRACT

Sialylation of glycoproteins and glycolipids is catalyzed by sialyltransferases in the Golgi of mammalian cells, whereby sialic acid residues are added at the nonreducing ends of oligosaccharides. Because sialylated glycans play critical roles in a number of human physio-pathological processes, the past two decades have witnessed the development of modified sialic acid derivatives for a better understanding of sialic acid biology and for the development of new therapeutic targets. However, nothing is known about how individual mammalian sialyltransferases tolerate and behave towards these unnatural CMP-sialic acid donors. In this study, we devised several approaches to investigate the donor specificity of the human ß-d-galactoside sialyltransferases ST6Gal I and ST3Gal I by using two CMP-sialic acids: CMP-Neu5Ac, and CMP-Neu5N-(4pentynoyl)neuraminic acid (CMP-SiaNAl), an unnatural CMP-sialic acid donor with an extended and functionalized N-acyl moiety.


Subject(s)
Antigens, CD/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Cytidine Monophosphate/analogs & derivatives , Glycolipids/metabolism , Glycoproteins/metabolism , Polysaccharides/metabolism , Sialic Acids/metabolism , Sialyltransferases/metabolism , Antigens, CD/chemistry , Antigens, CD/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cloning, Molecular , Cytidine Monophosphate/chemistry , Cytidine Monophosphate/metabolism , Cytidine Monophosphate N-Acetylneuraminic Acid/chemistry , Gene Expression , Glycolipids/chemistry , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , HEK293 Cells , Humans , Kinetics , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/chemistry , Neisseria meningitidis/enzymology , Polysaccharides/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sialic Acids/chemistry , Sialyltransferases/chemistry , Sialyltransferases/genetics , Substrate Specificity , beta-Galactoside alpha-2,3-Sialyltransferase
8.
Biochem J ; 473(13): 1905-16, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27114558

ABSTRACT

CMP-sialic acid synthetase (CSAS) is a key enzyme of the sialylation pathway. CSAS produces the activated sugar donor, CMP-sialic acid, which serves as a substrate for sialyltransferases to modify glycan termini with sialic acid. Unlike other animal CSASs that normally localize in the nucleus, Drosophila melanogaster CSAS (DmCSAS) localizes in the cell secretory compartment, predominantly in the Golgi, which suggests that this enzyme has properties distinct from those of its vertebrate counterparts. To test this hypothesis, we purified recombinant DmCSAS and characterized its activity in vitro Our experiments revealed several unique features of this enzyme. DmCSAS displays specificity for N-acetylneuraminic acid as a substrate, shows preference for lower pH and can function with a broad range of metal cofactors. When tested at a pH corresponding to the Golgi compartment, the enzyme showed significant activity with several metal cations, including Zn(2+), Fe(2+), Co(2+) and Mn(2+), whereas the activity with Mg(2+) was found to be low. Protein sequence analysis and site-specific mutagenesis identified an aspartic acid residue that is necessary for enzymatic activity and predicted to be involved in co-ordinating a metal cofactor. DmCSAS enzymatic activity was found to be essential in vivo for rescuing the phenotype of DmCSAS mutants. Finally, our experiments revealed a steep dependence of the enzymatic activity on temperature. Taken together, our results indicate that DmCSAS underwent evolutionary adaptation to pH and ionic environment different from that of counterpart synthetases in vertebrates. Our data also suggest that environmental temperatures can regulate Drosophila sialylation, thus modulating neural transmission.


Subject(s)
Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Amino Acid Sequence , Animals , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Cells, Cultured , Drosophila , Drosophila Proteins/classification , Drosophila Proteins/genetics , Humans , Hydrogen-Ion Concentration , Kinetics , Magnesium/metabolism , Manganese/metabolism , Mutation , N-Acylneuraminate Cytidylyltransferase/classification , N-Acylneuraminate Cytidylyltransferase/genetics , Phylogeny , Structure-Activity Relationship , Substrate Specificity , Temperature
9.
Glycobiology ; 26(11): 1151-1156, 2016 11.
Article in English | MEDLINE | ID: mdl-27543325

ABSTRACT

Natural and synthetically modified cytidine monophosphate activated sialic acids (CMP-Sias) are essential research assets in the field of glycobiology: among other applications, they can be used to probe glycans, detect sialylation defects at the cell surface or carry out detailed studies of sialyltransferase activities. However, these chemical tools are notoriously unstable because of hydrolytic decomposition, and are very time-consuming and costly to obtain. They are nigh impossible to store with satisfactory purity, and their preparation requires multiple laborious purification steps that usually lead to heavy product loss. Using in situ time-resolved 31P phosphorus nuclear magnetic resonance (31P NMR), we precisely established the kinetics of formation and degradation of a number of CMP-Sias including CMP-Neu5Ac, CMP-Neu5Gc, CMP-SiaNAl and CMP-SiaNAz in several experimental conditions. 31P NMR can be carried out in undeuterated solvents and is a sensitive and nondestructive technique that allows for direct in situ monitoring and optimization of chemo-enzymatic syntheses that involve phosphorus-containing species. Thus, we showed that CMP-sialic acid derivatives can be robustly obtained in high yields using the readily available Neisseria meningitidis CMP-sialic acid synthase. This integrated workflow takes less than an hour, and the freshly prepared CMP-Sias can be directly transferred to sialylation biological assays without any purification step.


Subject(s)
Cytidine Monophosphate/chemistry , Molecular Probes/chemistry , Polysaccharides/analysis , Sialic Acids/chemistry , Cytidine Monophosphate/biosynthesis , Cytidine Monophosphate/chemical synthesis , Molecular Probes/biosynthesis , Molecular Probes/chemical synthesis , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/enzymology , Sialic Acids/biosynthesis , Sialic Acids/chemical synthesis
10.
Org Biomol Chem ; 14(36): 8586-97, 2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27548611

ABSTRACT

A facile one-pot two-enzyme chemoenzymatic approach has been established for the gram (Neu4,5Ac2α3Lac, 1.33 g) and preparative scale (Neu4,5Ac2α3LNnT) synthesis of monotreme milk oligosaccharides. Other O-acetyl-5-N-acetylneuraminic acid (Neu4,5Ac2)- or 4-O-acetyl-5-N-glycolylneuraminic acid (Neu4Ac5Gc) -containing α2-3-sialosides have also been synthesized in the preparative scale. Used as an effective probe, Neu4,5Ac2α3GalßpNP was found to be a suitable substrate by human influenza A viruses but not bacterial sialidases.


Subject(s)
Milk/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Oligosaccharides/biosynthesis , Sialic Acids/biosynthesis , Sialyltransferases/metabolism , Animals , Milk/metabolism , Molecular Conformation , Oligosaccharides/chemistry , Sialic Acids/chemistry
11.
Top Curr Chem ; 366: 139-67, 2015.
Article in English | MEDLINE | ID: mdl-24141690

ABSTRACT

Sialoglycoconjugates form the outermost layer of animal cells and play a crucial role in cellular communication processes. An essential step in the biosynthesis of sialylated glycoconjugates is the activation of sialic acid to the monophosphate diester CMP-sialic acid. Only the activated sugar is transported into the Golgi apparatus and serves as a substrate for the linkage-specific sialyltransferases. Interference with sugar activation abolishes sialylation and is embryonic lethal in mammals. In this chapter we focus on the enzyme catalyzing the activation of sialic acid, the CMP-sialic acid synthetase (CMAS), and compare the enzymatic properties of CMASs isolated from different species. Information concerning the reaction mechanism and active site architecture is included. Moreover, the unusual nuclear localization of vertebrate CMASs as well as the biotechnological application of bacterial CMAS enzymes is addressed.


Subject(s)
Bacteria/enzymology , Cytidine Monophosphate N-Acetylneuraminic Acid/metabolism , Eukaryotic Cells/enzymology , Glycoconjugates/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Amino Acid Sequence , Animals , Bacteria/chemistry , Biological Transport , Catalytic Domain , Cell Communication , Cytidine Monophosphate N-Acetylneuraminic Acid/chemistry , Eukaryotic Cells/chemistry , Glycoconjugates/chemistry , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , N-Acylneuraminate Cytidylyltransferase/chemistry , Sequence Homology, Amino Acid , Species Specificity
12.
J Neurosci ; 33(30): 12306-15, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23884937

ABSTRACT

While sialylation plays important functions in the nervous system, the complexity of glycosylation pathways and limitations of genetic approaches preclude the efficient analysis of these functions in mammalian organisms. Drosophila has recently emerged as a promising model for studying neural sialylation. Drosophila sialyltransferase, DSiaT, was shown to be involved in the regulation of neural transmission. However, the sialylation pathway was not investigated in Drosophila beyond the DSiaT-mediated step. Here we focused on the function of Drosophila cytidine monophosphate-sialic acid synthetase (CSAS), the enzyme providing a sugar donor for DSiaT. Our results revealed that the expression of CSAS is tightly regulated and restricted to the CNS throughout development and in adult flies. We generated CSAS mutants and analyzed their phenotypes using behavioral and physiological approaches. Our experiments demonstrated that mutant phenotypes of CSAS are similar to those of DSiaT, including decreased longevity, temperature-induced paralysis, locomotor abnormalities, and defects of neural transmission at neuromuscular junctions. Genetic interactions between CSAS, DSiaT, and voltage-gated channel genes paralytic and seizure were consistent with the hypothesis that CSAS and DSiaT function within the same pathway regulating neural excitability. Intriguingly, these interactions also suggested that CSAS and DSiaT have some additional, independent functions. Moreover, unlike its mammalian counterparts that work in the nucleus, Drosophila CSAS was found to be a glycoprotein-bearing N-glycans and predominantly localized in vivo to the Golgi compartment. Our work provides the first systematic analysis of in vivo functions of a eukaryotic CSAS gene and sheds light on evolutionary relationships among metazoan CSAS proteins.


Subject(s)
Cytidine Monophosphate/metabolism , Drosophila Proteins/genetics , Drosophila/enzymology , Ligases/genetics , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , Nervous System Physiological Phenomena/genetics , Animals , Animals, Genetically Modified , Drosophila/genetics , Drosophila Proteins/metabolism , Evolution, Molecular , Gene Expression Regulation, Developmental/physiology , Ligases/metabolism , Longevity/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Paralysis/genetics , Paralysis/metabolism , Secretory Vesicles/physiology , Sialyltransferases/genetics , Sialyltransferases/metabolism , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Temperature
13.
Nanoscale ; 16(31): 14932-14939, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39046038

ABSTRACT

6'-Sialyllactose (6'-SL), the most abundant sialylated human milk oligosaccharide, has attracted attention for its potential application in supplementary infant formulas. Herein, we report a facile strategy to construct a cascade bioreactor for the enzymatic synthesis of 6'-SL by co-immobilizing an enzymatic module consisting of CMP-sialic acid synthase and α-2,6-sialyltransferase into hierarchically porous MIL-53 (HP-MIL-53). The as-prepared HP-MIL-53 showed high enzyme immobilization capacity, reaching 226 mg g-1. Furthermore, the co-immobilized enzymes exhibited higher initial catalytic efficiency, and thermal, pH and storage stability than the free ones. Finally, the 6'-SL yield remained >80% after 13 cycles of use. We expect that HP-MIL-53 would have potential industrial applications in the enzymatic modular synthesis of 6'-SL and other glycans.


Subject(s)
Enzymes, Immobilized , Sialyltransferases , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Sialyltransferases/metabolism , Porosity , Humans , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Oligosaccharides/biosynthesis , N-Acylneuraminate Cytidylyltransferase/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , Bioreactors , Milk, Human/chemistry , Milk, Human/metabolism , Lactose/chemistry , Lactose/analogs & derivatives , Lactose/metabolism , Hydrogen-Ion Concentration , beta-D-Galactoside alpha 2-6-Sialyltransferase
14.
J Biol Chem ; 287(16): 13239-48, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22351762

ABSTRACT

Sialic acids (Sia) form the nonreducing end of the bulk of cell surface-expressed glycoconjugates. They are, therefore, major elements in intercellular communication processes. The addition of Sia to glycoconjugates requires metabolic activation to CMP-Sia, catalyzed by CMP-Sia synthetase (CMAS). This highly conserved enzyme is located in the cell nucleus in all vertebrates investigated to date, but its nuclear function remains elusive. Here, we describe the identification and characterization of two Cmas enzymes in Danio rerio (dreCmas), one of which is exclusively localized in the cytosol. We show that the two cmas genes most likely originated from the third whole genome duplication, which occurred at the base of teleost radiation. cmas paralogues were maintained in fishes of the Otocephala clade, whereas one copy got subsequently lost in Euteleostei (e.g. rainbow trout). In zebrafish, the two genes exhibited a distinct spatial expression pattern. The products of these genes (dreCmas1 and dreCmas2) diverged not only with respect to subcellular localization but also in substrate specificity. Nuclear dreCmas1 favored N-acetylneuraminic acid, whereas the cytosolic dreCmas2 showed highest affinity for 5-deamino-neuraminic acid. The subcellular localization was confirmed for the endogenous enzymes in fractionated zebrafish lysates. Nuclear entry of dreCmas1 was mediated by a bipartite nuclear localization signal, which seemed irrelevant for other enzymatic functions. With the current demonstration that in zebrafish two subfunctionalized cmas paralogues co-exist, we introduce a novel and unique model to detail the roles that CMAS has in the nucleus and in the sialylation pathways of animal cells.


Subject(s)
Evolution, Molecular , N-Acylneuraminate Cytidylyltransferase/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Nucleus/enzymology , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Glycosylation , Mice , Molecular Sequence Data , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , NIH 3T3 Cells , RNA, Messenger/genetics , Substrate Specificity/physiology , Zebrafish/embryology
15.
Org Biomol Chem ; 11(5): 842-8, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23241945

ABSTRACT

Fluorinated Thomsen-Friedenreich (T) antigens were synthesized efficiently from chemically produced fluorinated monosaccharides using a highly efficient one-pot two-enzyme chemoenzymatic approach containing a galactokinase and a D-galactosyl-ß1-3-N-acetyl-D-hexosamine phosphorylase. These fluorinated T-antigens were further sialylated to form fluorinated ST-antigens using a one-pot two-enzyme system containing a CMP-sialic acid synthetase and an α-2-3-sialyltransferase.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/chemistry , Antigens, Tumor-Associated, Carbohydrate/metabolism , Mucins/chemistry , Mucins/metabolism , Antigens, Viral, Tumor/metabolism , Carbohydrate Sequence , Halogenation , Humans , Molecular Sequence Data , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/enzymology , Pasteurella multocida/enzymology , Sialyltransferases/metabolism , beta-Galactoside alpha-2,3-Sialyltransferase
16.
J Am Soc Nephrol ; 23(8): 1319-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22745475

ABSTRACT

The role of sialylation in kidney biology is not fully understood. The synthesis of sialoglycoconjugates, which form the outermost structures of animal cells, requires CMP-sialic acid, which is a product of the nuclear enzyme CMAS. We used a knock-in strategy to create a mouse with point mutations in the canonical nuclear localization signal of CMAS, which relocated the enzyme to the cytoplasm of transfected cells without affecting its activity. Although insufficient to prevent nuclear entry in mice, the mutation led to a drastically reduced concentration of nuclear-expressed enzyme. Mice homozygous for the mutation died from kidney failure within 72 hours after birth. The Cmas(nls) mouse exhibited podocyte foot process effacement, absence of slit diaphragms, and massive proteinuria, recapitulating features of nephrin-knockout mice and of patients with Finnish-type congenital nephrotic syndrome. Although the Cmas(nls) mouse displayed normal sialylation in all organs including kidney, a critical shortage of CMP-sialic acid prevented sialylation of nephrin and podocalyxin in the maturing podocyte where it is required during the formation of foot processes. Accordingly, the sialylation defects progressed with time and paralleled the morphologic changes. In summary, sialylation is critical during the development of the glomerular filtration barrier and required for the proper function of nephrin. Whether altered sialylation impairs nephrin function in human disease requires further study.


Subject(s)
Glomerular Filtration Barrier/embryology , Membrane Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , N-Acylneuraminate Cytidylyltransferase/metabolism , Podocytes/physiology , Animals , Cell Nucleus/metabolism , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , N-Acylneuraminate Cytidylyltransferase/genetics , Phenotype , Podocytes/ultrastructure , Sialoglycoproteins/metabolism
17.
Microbiol Spectr ; 11(6): e0294423, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37850751

ABSTRACT

IMPORTANCE: The Gram-negative coccobacillus Mannheimia haemolytica is a natural inhabitant of the upper respiratory tract in ruminants and the most common bacterial agent involved in bovine respiratory disease complex development. Key virulence factors harbored by M. haemolytica are leukotoxin, lipopolysaccharide, capsule, adhesins, and neuraminidase which are involved in evading innate and adaptive immune responses. In this study, we have shown that CMP-sialic acid synthetase (neuA) is necessary for the incorporation of sialic acid onto the membrane, and inactivation of neuA results in increased phagocytosis and complement-mediated killing of M. haemolytica, thus demonstrating that sialylation contributes to the virulence of M. haemolytica.


Subject(s)
Mannheimia haemolytica , Cattle , Animals , Mannheimia haemolytica/genetics , Mannheimia haemolytica/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Serogroup , Gene Deletion , Phagocytosis
18.
Glycobiology ; 22(4): 479-91, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22057003

ABSTRACT

Sialic acid, a common terminal substitution of glycoconjugates, has been so far consistently identified in all vertebrates as well as in a growing number of bacterial species. It is assumed to be widely distributed among animal species of the deuterostome phylum, based on its identification in few echinoderm and all vertebrate species. However, whole sections of deuterostome, especially those intermediate species between invertebrates and vertebrates including cephalochordates, urochordates and hemichordates, are still unexplored in term of sialylation capacities. The discovery of functional sialic acid machinery in some of these species may shed new light onto the evolution of glycosylation capacities in deuterostome lineage. In a first approach, we investigated the sialylation pattern of a cephalocordate species, Branchiostoma belcheri, which occupies a strategic phylogenetic position to understand the transition of invertebrates toward vertebrates. Structural analysis of B. belcheri glycoconjugates established that this organism synthesizes large quantities of various sialic acids, some of which present rare or novel structures such as methylated sialic acids. These sialic acids were shown to be mainly associated with mono- and disialylated core 1-type O-glycans. Moreover, screening of the animal organs revealed the existence of exquisite tissue specificity in the distribution of sialic acids. Description of sialylation profiles was then correlated with the expression patterns of key enzymes involved in the biosynthesis of major forms of sialic acids, which provides the first complete overview of the sialylation patterns in cephalochordates.


Subject(s)
Chordata, Nonvertebrate/metabolism , Sialic Acids/metabolism , Animals , Biological Evolution , Carbohydrate Conformation , Chordata, Nonvertebrate/enzymology , Chordata, Nonvertebrate/genetics , Female , Glycolipids/metabolism , Glycomics , Glycoproteins/metabolism , Glycosylation , Male , Methylation , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , N-Acylneuraminate Cytidylyltransferase/genetics , N-Acylneuraminate Cytidylyltransferase/metabolism , Organ Specificity , Ovary/metabolism , Polysaccharides/metabolism , Sialic Acids/isolation & purification , Sialyltransferases/genetics , Sialyltransferases/metabolism , Sugar Acids/metabolism , Testis/metabolism , Transcription, Genetic , Vertebrates/genetics , beta-Galactoside alpha-2,3-Sialyltransferase
19.
Appl Microbiol Biotechnol ; 93(6): 2411-23, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21968653

ABSTRACT

Cytidine 5'-monophosphate (CMP)-sialic acid synthetases (CSSs) catalyze the formation of CMP-sialic acid from CTP and sialic acid, a key step for sialyltransferase-catalyzed biosynthesis of sialic acid-containing oligosaccharides and glycoconjugates. More than 50 different sialic acid forms have been identified in nature. To facilitate the enzymatic synthesis of sialosides with diverse naturally occurring sialic acid forms and their non-natural derivatives, CMP-sialic acid synthetases with promiscuous substrate specificity are needed. Herein we report the cloning, characterization, and substrate specificity studies of a new CSS from Pasteurella multocida strain P-1059 (PmCSS) and a CSS from Haemophillus ducreyi (HdCSS). Based on protein sequence alignment and substrate specificity studies of these two CSSs and a Neisseria meningitidis CSS (NmCSS), as well as crystal structure modeling and analysis of NmCSS, NmCSS mutants (NmCSS_S81R and NmCSS_Q163A) with improved substrate promiscuity were generated. The strategy of combining substrate specificity studies of enzymes from different sources and protein crystal structure studies can be a general approach for designing enzyme mutants with improved activity and substrate promiscuity.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , N-Acylneuraminate Cytidylyltransferase/chemistry , N-Acylneuraminate Cytidylyltransferase/metabolism , Neisseria meningitidis/enzymology , Pasteurella multocida/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Haemophilus ducreyi/chemistry , Haemophilus ducreyi/enzymology , Haemophilus ducreyi/genetics , Molecular Sequence Data , Mutation , N-Acylneuraminate Cytidylyltransferase/genetics , Neisseria meningitidis/chemistry , Neisseria meningitidis/genetics , Pasteurella multocida/chemistry , Pasteurella multocida/genetics , Sequence Alignment , Substrate Specificity
20.
Biosci Biotechnol Biochem ; 75(1): 47-53, 2011.
Article in English | MEDLINE | ID: mdl-21228490

ABSTRACT

A cytidine 5'-monophospho-N-acetylneuraminic acid (CMP-Neu5Ac) synthetase was found in a crude extract prepared from Photobacterium leiognathi JT-SHIZ-145, a marine bacterium that also produces a ß-galactoside α2,6-sialyltransferase. The CMP-Neu5Ac synthetase was purified from the crude extract of the cells by a combination of anion-exchange and gel filtration column chromatography. The purified enzyme migrated as a single band (60 kDa) on sodium dodecylsulfate-polyacrylamide gel electrophoresis. The activity of the enzyme was maximal at 35 °C at pH 9.0, and the synthetase required Mg(2+) for activity. Although these properties are similar to those of other CMP-Neu5Ac synthetases isolated from bacteria, this synthetase produced not only CMP-Neu5Ac from cytidine triphosphate and Neu5Ac, but also CMP-N-glycolylneuraminic acid from cytidine triphosphate and N-glycolylneuraminic acid, unlike CMP-Neu5Ac synthetase purified from Escherichia coli.


Subject(s)
N-Acylneuraminate Cytidylyltransferase/isolation & purification , Photobacterium/enzymology , Kinetics , N-Acylneuraminate Cytidylyltransferase/biosynthesis , N-Acylneuraminate Cytidylyltransferase/metabolism , Oligosaccharides/analysis , Oligosaccharides/biosynthesis , Oligosaccharides/isolation & purification , Photobacterium/classification , Photobacterium/metabolism , Sialyltransferases/metabolism , Substrate Specificity , beta-D-Galactoside alpha 2-6-Sialyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL