Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 300(5): 107229, 2024 May.
Article in English | MEDLINE | ID: mdl-38537698

ABSTRACT

Mucosal-associated invariant T (MAIT) cells can elicit immune responses against riboflavin-based antigens presented by the evolutionary conserved MHC class I related protein, MR1. While we have an understanding of the structural basis of human MAIT cell receptor (TCR) recognition of human MR1 presenting a variety of ligands, how the semi-invariant mouse MAIT TCR binds mouse MR1-ligand remains unknown. Here, we determine the crystal structures of 2 mouse TRAV1-TRBV13-2+ MAIT TCR-MR1-5-OP-RU ternary complexes, whose TCRs differ only in the composition of their CDR3ß loops. These mouse MAIT TCRs mediate high affinity interactions with mouse MR1-5-OP-RU and cross-recognize human MR1-5-OP-RU. Similarly, a human MAIT TCR could bind mouse MR1-5-OP-RU with high affinity. This cross-species recognition indicates the evolutionary conserved nature of this MAIT TCR-MR1 axis. Comparing crystal structures of the mouse versus human MAIT TCR-MR1-5-OP-RU complexes provides structural insight into the conserved nature of this MAIT TCR-MR1 interaction and conserved specificity for the microbial antigens, whereby key germline-encoded interactions required for MAIT activation are maintained. This is an important consideration for the development of MAIT cell-based therapeutics that will rely on preclinical mouse models of disease.


Subject(s)
Histocompatibility Antigens Class I , Minor Histocompatibility Antigens , Mucosal-Associated Invariant T Cells , Ribitol , Animals , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/chemistry , Minor Histocompatibility Antigens/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/immunology , Minor Histocompatibility Antigens/chemistry , Mice , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Humans , Ribitol/analogs & derivatives , Ribitol/metabolism , Ribitol/chemistry , Uracil/analogs & derivatives , Uracil/metabolism , Uracil/chemistry , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Crystallography, X-Ray
2.
Sci Adv ; 10(9): eadj3864, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38416829

ABSTRACT

Wall teichoic acid (WTA), a covalent adduct of Gram-positive bacterial cell wall peptidoglycan, contributes directly to virulence and antibiotic resistance in pathogenic species. Polymerization of the Staphylococcus aureus WTA ribitol-phosphate chain is catalyzed by TarL, a member of the largely uncharacterized TagF-like family of membrane-associated enzymes. We report the cryo-electron microscopy structure of TarL, showing a tetramer that forms an extensive membrane-binding platform of monotopic helices. TarL is composed of an amino-terminal immunoglobulin-like domain and a carboxyl-terminal glycosyltransferase-B domain for ribitol-phosphate polymerization. The active site of the latter is complexed to donor substrate cytidine diphosphate-ribitol, providing mechanistic insights into the catalyzed phosphotransfer reaction. Furthermore, the active site is surrounded by electropositive residues that serve to retain the lipid-linked acceptor for polymerization. Our data advance general insight into the architecture and membrane association of the still poorly characterized monotopic membrane protein class and present molecular details of ribitol-phosphate polymerization that may aid in the design of new antimicrobials.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Staphylococcus aureus/metabolism , Cryoelectron Microscopy , Methicillin-Resistant Staphylococcus aureus/metabolism , Virulence , Ribitol/metabolism , Teichoic Acids/analysis , Teichoic Acids/chemistry , Teichoic Acids/metabolism , Phosphates/metabolism , Drug Resistance, Microbial
3.
J Biochem ; 175(4): 418-425, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38140954

ABSTRACT

The core M3 O-mannosyl glycan on α-dystroglycan serves as the binding epitope for extracellular matrix molecules. Defects in core M3 glycans cause congenital muscular dystrophies that are collectively known as dystroglycanopathies. The core M3 glycan contains a tandem D-ribitol-5-phosphate (Rbo5P) structure, which is synthesized by the Rbo5P-transferases fukutin and fukutin-related protein using CDP-ribitol (CDP-Rbo) as a donor substrate. CDP-Rbo is synthesized from CTP and Rbo5P by CDP-Rbo pyrophosphorylase A. However, the Rbo5P biosynthesis pathway has yet to be elucidated in mammals. Here, we investigated the reductase activities toward four substrates, including ribose, ribulose, ribose-phosphate and ribulose-phosphate, to identify the intracellular Rbo5P production pathway and elucidated the role of the aldo-keto reductases AKR1A1, AKR1B1 and AKR1C1 in those pathways. It was shown that the ribose reduction pathway is the endogenous pathway that contributes most to Rbo5P production in HEK293T cells and that AKR1B1 is the major reductase in this pathway.


Subject(s)
Ribitol , Ribose , Humans , Animals , Ribitol/metabolism , Phosphates , HEK293 Cells , Dystroglycans/metabolism , Oxidoreductases , Mammals , Polysaccharides/metabolism , Aldehyde Reductase
4.
Mol Ther ; 31(12): 3478-3489, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37919902

ABSTRACT

Mutations in the fukutin-related protein (FKRP) gene cause dystroglycanopathy, with disease severity ranging from mild LGMD2I to severe congenital muscular dystrophy. Recently, considerable progress has been made in developing experimental therapies, with adeno-associated virus (AAV) gene therapy and ribitol treatment demonstrating significant therapeutic effect. However, each treatment has its strengths and weaknesses. AAV gene therapy can achieve normal levels of transgene expression, but it requires high doses, with toxicity concerns and variable distribution. Ribitol relies on residual FKRP function and restores limited levels of matriglycan. We hypothesized that these two treatments can work synergistically to offer an optimized therapy with efficacy and safety unmatched by each treatment alone. The most effective treatment is the combination of high-dose (5e-13 vg/kg) AAV-FKRP with ribitol, whereas low dose (1e-13 vg/kg) AAV-FKRP combined with ribitol showed a 22.6% increase in positive matriglycan fibers and the greater improvement in pathology when compared to low-dose AAV-FKRP alone. Together, our results support the potential benefits of combining ribitol with AAV gene therapy for treating FKRP-related muscular dystrophy. The fact that ribitol is a metabolite in nature and has already been tested in animal models and clinical trials in humans without severe side effects provides a safety profile for it to be trialed in combination with AAV gene therapy.


Subject(s)
Muscular Dystrophies , Pentosyltransferases , Animals , Humans , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Pentosyltransferases/therapeutic use , Ribitol/metabolism , Ribitol/therapeutic use , Dependovirus/genetics , Dependovirus/metabolism , Dystroglycans/metabolism , Muscular Dystrophies/drug therapy , Genetic Therapy/methods , Mutation , Muscle, Skeletal/metabolism
5.
J Biochem ; 173(5): 333-335, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-36760122

ABSTRACT

Dystroglycan (DG), a muscular transmembrane protein, plays a critical role in transducing extracellular matrix-derived signals to the cytoskeleton and provides physical strength to skeletal muscle cell membranes. The extracellular domain of DG, α-DG, displays unique glycosylation patterns. Fully functional glycosylation is required for this domain to interact with components of extracellular matrices, including laminin. One of the unique sugar compositions found in such functional glycans on DG is two ribitol phosphates that are transferred by the sequential actions of fukutin (FKTN) and fukutin-related protein (FKRP), which use CDP-ribitol as a donor substrate. These are then further primed for matriglycan biosynthesis. A recent in vitro study reported that glycerol phosphate could be similarly added to α-DG by FKTN and FKRP if they used CDP-glycerol (CDP-Gro) as a donor substrate. However, the physiological relevance of these findings remains elusive. Imae et al. addressed the knowledge gap regarding whether CDP-Gro is present in mammals and how CDP-Gro is synthesized and functions in mammals.


Subject(s)
Dystroglycans , Pentosyltransferases , Animals , Dystroglycans/metabolism , Glycerol , Glycosylation , Pentosyltransferases/metabolism , Ribitol/metabolism , Ribitol/pharmacology
6.
ACS Chem Biol ; 17(6): 1513-1523, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35670527

ABSTRACT

Ribitol phosphate modifications to the core M3 O-mannosyl glycan are important for the functional maturation of α-dystroglycan. Three sequentially extended partial structures of the core M3 O-mannosyl glycan including a tandem ribitol phosphate were regio- and stereo-selectively synthesized: Rbo5P-3GalNAcß, Rbo5P-1Rbo5P-3GalNAcß, and Xylß1-4Rbo5P-1Rbo5P-3GalNAcß (Rbo5P, d-ribitol-5-phosphate; GalNAc, N-acetyl-d-galactosamine; Xyl, d-xylose). Rbo5P-3GalNAcß with p-nitrophenyl at the aglycon part served as a substrate for ribitol phosphate transferase (FKRP, fukutin-related protein), and its product was glycosylated by the actions of a series of glycosyltransferases, namely, ribitol xylosyltransferase 1 (RXYLT1), ß1,4-glucuronyltransferase 1 (B4GAT1), and like-acetyl-glucosaminyltransferase (LARGE). Rbo5P-3GalNAcß equipped with an alkyne-type aglycon was also active for FKRP. The molecular information obtained on FKRP suggests that Rbo5P-3GalNAcß derivatives are the minimal units required as the acceptor glycan for Rbo5P transfer and may serve as a precursor for the elongation of the core M3 O-mannosyl glycan.


Subject(s)
Phosphates , Ribitol , Dystroglycans/chemistry , Dystroglycans/metabolism , Glycosylation , Pentosyltransferases/metabolism , Polysaccharides/metabolism , Ribitol/metabolism
7.
Microbiol Spectr ; 10(2): e0033622, 2022 04 27.
Article in English | MEDLINE | ID: mdl-35311554

ABSTRACT

Tetragenococcus halophilus, a halophilic lactic acid bacterium, is used in the fermentation process of soy sauce manufacturing. For many years, bacteriophage infections of T. halophilus have been a major industrial problem that causes fermentation failure. However, studies focusing on the mechanisms of tetragenococcal host-phage interactions are not sufficient. In this study, we generated two phage-insensitive derivatives from the parental strain T. halophilus WJ7, which is susceptible to the virulent phage phiWJ7. Whole-genome sequencing of the derivatives revealed that insertion sequences were transposed into a gene encoding poly(ribitol phosphate) polymerase (TarL) in both derivatives. TarL is responsible for the biosynthesis of ribitol-containing wall teichoic acid, and WJ7 was confirmed to contain ribitol in extracted wall teichoic acid, but the derivative was not. Cell walls of WJ7 irreversibly adsorbed phiWJ7, but those of the phage-insensitive derivatives did not. Additionally, 25 phiWJ7-insensitive derivatives were obtained, and they showed mutations not only in tarL but also in tarI and tarJ, which are responsible for the synthesis of CDP-ribitol. These results indicate that phiWJ7 targets the ribitol-containing wall teichoic acid of host cells as a binding receptor. IMPORTANCE Information about the mechanisms of host-phage interactions is required for the development of efficient strategies against bacteriophage infections. Here, we identified the ribitol-containing wall teichoic acid as a host receptor indispensable for bacteriophage infection. The complete genome sequence of tetragenococcal phage phiWJ7 belonging to the family Rountreeviridae is also provided here. This study could become the foundation for a better understanding of host-phage interactions of tetragenococci.


Subject(s)
Bacteriophages , Ribitol , Bacteriophages/genetics , Cell Wall/metabolism , Enterococcaceae/metabolism , Ribitol/metabolism
8.
Elife ; 102021 01 29.
Article in English | MEDLINE | ID: mdl-33513091

ABSTRACT

Mutations in the fukutin-related protein (FKRP) cause Walker-Warburg syndrome (WWS), a severe form of congenital muscular dystrophy. Here, we established a WWS human induced pluripotent stem cell-derived myogenic model that recapitulates hallmarks of WWS pathology. We used this model to investigate the therapeutic effect of metabolites of the pentose phosphate pathway in human WWS. We show that functional recovery of WWS myotubes is promoted not only by ribitol but also by its precursor ribose. Moreover, we found that the combination of each of these metabolites with NAD+ results in a synergistic effect, as demonstrated by rescue of α-dystroglycan glycosylation and laminin binding capacity. Mechanistically, we found that FKRP residual enzymatic capacity, characteristic of many recessive FKRP mutations, is required for rescue as supported by functional and structural mutational analyses. These findings provide the rationale for testing ribose/ribitol in combination with NAD+ to treat WWS and other diseases associated with FKRP mutations.


Healthy muscles are complex machines that require a myriad of finely tuned molecules to work properly. For instance, a protein called alpha-DG sits at the surface of healthy muscle cells, where it strengthens the tissue by latching onto other proteins in the environment. To perform its role correctly, it first needs to be coated with sugar molecules, a complex process which requires over 20 proteins, including the enzyme FKRP. Faulty forms of FKRP reduce the number of sugars added to alpha-DG, causing the muscle tissue to weaken and waste away, potentially leading to severe forms of diseases known as muscular dystrophies. Drugs that can restore alpha-DG sugar molecules could help to treat these conditions. Previous studies on mice and fish have highlighted two potential candidates, known as ribitol and NAD+, which can help to compensate for reduced FKRP activity and allow sugars to be added to alpha-DG again. Yet no model is available to test these molecules on actual human muscle cells. Here, Ortiz-Cordero et al. developed such a model in the laboratory by growing muscle cells from naïve, undifferentiated cells generated from skin given by a muscular dystrophy patient with a faulty form of FKRP. The resulting muscle fibers are in essence identical to the ones present in the individual. As such, they can help to understand the effect various drugs have on muscular dystrophies. The cells were then put in contact with either NAD+, ribitol, or a precursor of ribitol known as ribose. Ortiz-Cordero et al. found that ribitol and ribose restored the ability of FKRP to add sugars to alpha-DG, reducing muscle damage. Combining NAD+ with ribitol or ribose had an even a bigger impact, further increasing the number of sugars on alpha-DG. The human muscle cell model developed by Ortiz-Cordero et al. could help to identify new compounds that can treat muscular conditions. In particular, the findings point towards NAD+, ribose and ribitol as candidates for treating FKRP-related muscular dystrophies. Further safety studies are now needed to evaluate whether these compounds could be used in patients.


Subject(s)
Dystroglycans/metabolism , Muscle Fibers, Skeletal/metabolism , NAD/pharmacology , Ribitol/metabolism , Ribose/metabolism , Cell Line , Glycosylation , Humans , Mutation , Pentosyltransferases/genetics
9.
Chembiochem ; 22(4): 672-678, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33034934

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are an abundant subset of innate-like T lymphocytes. MAIT cells are activated by microbial riboflavin-derived antigens, such as 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU), when presented by the major histocompatibility complex (MHC) class I-related protein (MR1). We have synthesized all stereoisomers of 5-OP-RU to investigate the effects of its stereochemistry on the MR1-dependent MAIT cell activation and MR1 upregulation. The analysis of MAIT cell activation by these 5-OP-RU isomers revealed that the stereocenters at the 2'- and 3'-OH groups in the ribityl tail are crucial for the recognition of MAIT-TCR, whereas that of 4'-OH group does not significantly affect the regulation of MAIT cell activity. Furthermore, kinetic analysis of complex formation between the ligands and MR1 suggested that 5-OP-RU forms a covalent bond to MR1 in cells within 1 hour. These findings provide guidelines for designing ligands that regulate MAIT cell functions.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens/metabolism , Mucosal-Associated Invariant T Cells/metabolism , Ribitol/analogs & derivatives , Uracil/analogs & derivatives , Humans , Kinetics , Ligands , Lymphocyte Activation , Ribitol/chemistry , Ribitol/metabolism , Stereoisomerism , Structure-Activity Relationship , Uracil/chemistry , Uracil/metabolism
10.
J Biol Chem ; 295(42): 14445-14457, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32817339

ABSTRACT

MR1 presents vitamin B-related metabolites to mucosal associated invariant T (MAIT) cells, which are characterized, in part, by the TRAV1-2+ αß T cell receptor (TCR). In addition, a more diverse TRAV1-2- MR1-restricted T cell repertoire exists that can possess altered specificity for MR1 antigens. However, the molecular basis of how such TRAV1-2- TCRs interact with MR1-antigen complexes remains unclear. Here, we describe how a TRAV12-2+ TCR (termed D462-E4) recognizes an MR1-antigen complex. We report the crystal structures of the unliganded D462-E4 TCR and its complex with MR1 presenting the riboflavin-based antigen 5-OP-RU. Here, the TRBV29-1 ß-chain of the D462-E4 TCR binds over the F'-pocket of MR1, whereby the complementarity-determining region (CDR) 3ß loop surrounded and projected into the F'-pocket. Nevertheless, the CDR3ß loop anchored proximal to the MR1 A'-pocket and mediated direct contact with the 5-OP-RU antigen. The D462-E4 TCR footprint on MR1 contrasted that of the TRAV1-2+ and TRAV36+ TCRs' docking topologies on MR1. Accordingly, diverse MR1-restricted T cell repertoire reveals differential docking modalities on MR1, thus providing greater scope for differing antigen specificities.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Amino Acid Sequence , Antigen Presentation , Binding Sites , Crystallography, X-Ray , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Humans , Minor Histocompatibility Antigens/chemistry , Minor Histocompatibility Antigens/genetics , Molecular Docking Simulation , Protein Refolding , Protein Structure, Tertiary , Receptors, Antigen, T-Cell, alpha-beta/chemistry , Receptors, Antigen, T-Cell, alpha-beta/genetics , Ribitol/analogs & derivatives , Ribitol/chemistry , Ribitol/metabolism , Surface Plasmon Resonance , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Uracil/analogs & derivatives , Uracil/chemistry , Uracil/metabolism
11.
PLoS One ; 15(7): e0235718, 2020.
Article in English | MEDLINE | ID: mdl-32639976

ABSTRACT

Sugar alcohols (polyols) are abundant carbohydrates in lichen-forming algae and transported to other lichen symbionts, fungi, and bacteria. Particularly, ribitol is an abundant polyol in the lichen Cetraria sp. Polyols have important physiological roles in lichen symbiosis, but polyol utilization in lichen-associated bacteria has been largely unreported. Herein, we purified and characterized a novel ribitol dehydrogenase (RDH) from a Cetraria sp.-associated bacterium Sphingomonas sp. PAMC 26621 grown on a minimal medium containing D-ribitol (the RDH hereafter referred to as SpRDH). SpRDH is present as a trimer in its native form, and the molecular weight of SpRDH was estimated to be 39 kDa by SDS-PAGE and 117 kDa by gel filtration chromatography. SpRDH converted D-ribitol to D-ribulose using NAD+ as a cofactor. As far as we know, SpRDH is the first RDH belonging to the medium-chain dehydrogenase/reductase family. Multiple sequence alignments indicated that the catalytic amino acid residues of SpRDH consist of Cys37, His65, Glu66, and Glu157, whereas those of short-chain RDHs consist of Ser, Tyr, and Lys. Furthermore, unlike other short-chain RDHs, SpRDH did not require divalent metal ions for its catalytic activity. Despite SpRDH originating from a psychrophilic Arctic bacterium, Sphingomonas sp., it had maximum activity at 60°C and exhibited high thermal stability within the 4-50°C range. Further studies on the structure/function relationship and catalytic mechanism of SpRDH will expand our understanding of its role in lichen symbiosis.


Subject(s)
Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Lichens/microbiology , Ribitol/metabolism , Sphingomonas/enzymology , Sugar Alcohol Dehydrogenases/isolation & purification , Sugar Alcohol Dehydrogenases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Sequence Homology , Sphingomonas/growth & development , Substrate Specificity , Sugar Alcohol Dehydrogenases/genetics
12.
Food Funct ; 11(7): 5782-5787, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32618294

ABSTRACT

Methylglyoxal (MGO) is the main antimicrobial determinant associated with using Manuka Honey as a topical dressing. While direct mechanisms of Manuka honey MGO's antimicrobial activity have been demonstrated, such as disruption of bacterial fimbria and flagella, no interaction of Manuka honey-derived MGO with antimicrobial effector cells of the immune system, such as mucosal-associated invariant T cells (MAIT cells), has yet been reported. MAIT cells are an abundant subset of human T cells, critical for regulating a diverse range of immune functions, including antimicrobial defense mechanisms but also mucosal barrier integrity. MAIT cells become activated by recognition of an important microbial metabolite, 5-amino-6-d-ribitylaminouracil (5-A-RU), which is produced by a wide range of microbial pathogens and commensals. Recognition is afforded when 5-A-RU condenses with mammalian-cell derived MGO to form the potent MAIT cell activator, 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU). Formation of 5-OP-RU and its subsequent presentation to MAIT cells by major histocompatibility (MHC)-related molecule 1 (MR1) facilitates host-pathogen and host-commensal interactions. While MGO is a metabolite naturally present in mammalian cells, it is unclear whether exogenous dietary MGO sources, such as those obtained from Manuka honey intake, can contribute to 5-OP-RU formation and enhance MAIT cell activation. In this work, we report that endogenous MGO is the rate-limiting substrate for converting microbial 5-A-RU to 5-OP-RU and that Manuka honey-derived MGO significantly enhances MAIT cell activation in vitro. Our findings posit a novel mechanism by which intake of a food item, such as Manuka honey, can potentially support immune homeostasis by enhancing MAIT cell-specific microbial sensing.


Subject(s)
Honey , Immunologic Factors/pharmacology , Leptospermum , Lymphocyte Activation/drug effects , Mucosal-Associated Invariant T Cells/metabolism , Pyruvaldehyde/pharmacology , Anti-Bacterial Agents/pharmacology , Apitherapy , Humans , Pyruvaldehyde/metabolism , Ribitol/analogs & derivatives , Ribitol/metabolism , Uracil/analogs & derivatives , Uracil/metabolism
13.
Appl Microbiol Biotechnol ; 104(13): 5663-5672, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32372201

ABSTRACT

Currently, due to the special functions and potential application values, rare sugars become the hot topic in carbohydrate fields. L-Ribulose, an isomer of L-ribose, is an expensive rare ketopentose. As an important precursor for other rare sugars and L-nucleoside analogue synthesis, L-ribulose attracts more and more attention in recent days. Compared with complicated chemical synthesis, the bioconversion method becomes a good alternative approach to L-ribulose production. Generally, the bioconversion of L-ribulose was linked with ribitol, L-arabinose, L-ribose, L-xylulose, and L-arabitol. Herein, an overview of recent advances in the metabolic pathway, chemical synthesis, bioproduction of L-ribulose, and the potential application of L-ribulose is reviewed in detail in this paper. KEY POINTS: 1. L-Ribulose is a rare sugar and the key precursor for L-ribose production. 2. L-Ribulose is the starting material for L-nucleoside derivative synthesis. 3. Chemical synthesis, bioproduction, and applications of L-ribulose are reviewed.


Subject(s)
Pentoses/metabolism , Arabinose/metabolism , Bacteria/classification , Bacteria/metabolism , Bacterial Proteins/metabolism , Biocatalysis , Biotransformation , Metabolic Networks and Pathways , Pentoses/chemical synthesis , Ribitol/metabolism , Ribose/metabolism , Sugar Alcohols/metabolism , Xylulose/metabolism
14.
Sci Rep ; 10(1): 4935, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188898

ABSTRACT

The laminin-binding glycan (matriglycan) on α-dystroglycan (α-DG) enables diverse roles, from neuronal development to muscle integrity. Reduction or loss of matriglycan has also been implicated in cancer development and metastasis, and specifically associated with high-grade tumors and poor prognoses in breast cancers. Hyperglycosylation of α-DG with LARGE overexpression is shown to inhibit cancer cell growth and tumorigenicity. We recently demonstrated that ribitol, considered to be a metabolic end-product, enhances matriglycan expression in dystrophic muscles in vivo. In the current study, we tested the hypothesis that ribitol could also enhance matriglycan expression in cancer cells. Our results showed for the first time that ribitol is able to significantly enhance the expression of matriglycan on α-DG in breast cancer cells. The ribitol effect is associated with an increase in levels of CDP-ribitol, the substrate for the ribitol-5-phosphate transferases FKRP and FKTN. Direct use of CDP-ribitol is also effective for matriglycan expression. Ribitol treatment does not alter the expression of FKRP, FKTN as well as LARGEs and ISPD which are critical for the synthesis of matriglycan. The results suggest that alteration in substrates could also be involved in regulation of matriglycan expression. Interestingly, expression of matriglycan is related to cell cycle progression with highest levels in S and G2 phases and ribitol treatment does not alter the pattern. Although matriglycan up-regulation does not affect cell cycle progression and proliferation of the cancer cells tested, the novel substrate-mediated treatment opens a new approach easily applicable to experimental systems in vivo for further exploitation of matriglycan expression in cancer progression and for therapeutic potential.


Subject(s)
Breast Neoplasms/metabolism , Dystroglycans/metabolism , Ribitol/metabolism , Breast Neoplasms/genetics , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chromatography, Liquid , Female , Gene Expression Regulation, Neoplastic , Glycosylation/drug effects , Humans , Immunohistochemistry , Pentosephosphates/metabolism , Ribitol/pharmacology , Tandem Mass Spectrometry
15.
Nat Commun ; 11(1): 303, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949166

ABSTRACT

α-Dystroglycan (α-DG) is a highly-glycosylated surface membrane protein. Defects in the O-mannosyl glycan of α-DG cause dystroglycanopathy, a group of congenital muscular dystrophies. The core M3 O-mannosyl glycan contains tandem ribitol-phosphate (RboP), a characteristic feature first found in mammals. Fukutin and fukutin-related protein (FKRP), whose mutated genes underlie dystroglycanopathy, sequentially transfer RboP from cytidine diphosphate-ribitol (CDP-Rbo) to form a tandem RboP unit in the core M3 glycan. Here, we report a series of crystal structures of FKRP with and without donor (CDP-Rbo) and/or acceptor [RboP-(phospho-)core M3 peptide] substrates. FKRP has N-terminal stem and C-terminal catalytic domains, and forms a tetramer both in crystal and in solution. In the acceptor complex, the phosphate group of RboP is recognized by the catalytic domain of one subunit, and a phosphate group on O-mannose is recognized by the stem domain of another subunit. Structure-based functional studies confirmed that the dimeric structure is essential for FKRP enzymatic activity.


Subject(s)
Muscular Dystrophies/metabolism , Nucleoside Diphosphate Sugars/chemistry , Nucleoside Diphosphate Sugars/metabolism , Pentosyltransferases/chemistry , Pentosyltransferases/metabolism , Catalytic Domain , Crystallography, X-Ray , Glycopeptides , HEK293 Cells , Humans , Models, Molecular , Muscular Dystrophies/genetics , Pentosyltransferases/genetics , Phosphates/metabolism , Polysaccharides/metabolism , Protein Conformation , Protein Domains , Ribitol/metabolism
16.
ACS Chem Biol ; 15(2): 437-445, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31909966

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are antibacterial effector T cells that react to pyrimidines derived from bacterial riboflavin synthesis presented by the monomorphic molecule MR1. A major challenge in MAIT cell research is that the commonly used MAIT agonist precursor, 5-amino-6-d-ribitylaminouracil (5-A-RU), is labile to autoxidation, resulting in a loss of biological activity. Here, we characterize two independent autoxidation processes by LCMS. To overcome the marked instability, we report the synthesis of a 5-A-RU prodrug generated by modification of the 5-amino group with a cleavable valine-citrulline-p-aminobenzyl carbamate. The compound is stable in prodrug form, with the parent amine (i.e., 5-A-RU) released only after enzymatic cleavage. Analysis of the prodrug in vitro and in vivo showed an enhanced MAIT cell activation profile compared to 5-A-RU, which was associated with preferential loading within recycling endosomes, a route used by some natural agonists. This prodrug design therefore overcomes the difficulties associated with 5-A-RU in biological studies and provides an opportunity to explore different presentation pathways.


Subject(s)
Endosomes/metabolism , Histocompatibility Antigens Class I/metabolism , Immunologic Factors/pharmacology , Lymphocyte Activation/drug effects , Minor Histocompatibility Antigens/metabolism , Mucosal-Associated Invariant T Cells/drug effects , Prodrugs/pharmacology , Animals , Humans , Immunologic Factors/chemical synthesis , Immunologic Factors/metabolism , Mice , Prodrugs/chemical synthesis , Prodrugs/metabolism , Ribitol/analogs & derivatives , Ribitol/chemical synthesis , Ribitol/metabolism , Ribitol/pharmacology , Uracil/analogs & derivatives , Uracil/chemical synthesis , Uracil/metabolism , Uracil/pharmacology
17.
Cryobiology ; 85: 39-46, 2018 12.
Article in English | MEDLINE | ID: mdl-30292810

ABSTRACT

The soil microalgae of the genus Heterococcus are found in cold environments and have been reported for the terrestrial ecosystems of several Sub-Antarctic and Antarctic Islands. This study focused on resistance of Heterococcus sp. to sub-zero temperature. Heterococcus sp. was isolated from soil samples from James Ross Island, Antarctica. Culture of Heterococcus sp. grown in liquid medium were used to study ribitol effects at sub-zero temperatures on the species resistance to rapid freezing (RF, immersion of a sample into liquid nitrogen) and consequent cultivation on agar. Before the experiment, Heterococcus sp. was cultured in liquid medium for 11 months and then treated in ribitol concentrations of 32 or 50 mM for 2 h. Then, 1 ml samples were frozen to -196 °C in liquid nitrogen (day 0) and inoculated on BBM agar after thawing. Number of living and dead cells was evaluated and the cell viability (Pν) was calculated repeatedly using the optical microscopy approach. The addition of ribitol caused a noticable increase in Pν on days 9, 12, 14 (with a Pν of 25-45% in ribitol-treated samples compared to 10% in the untreated control). In the following period (d 16-19), the positive effect of ribitol on Pν was less pronounced but still statistically significant. To evaluate the negative effects of RF on chlorophyll fluorescence parameters, the potential yield of photochemical reactions in PS II (FV/FM), and the effective quantum yield of photochemical reactions in PS II (ФPSII) were measured immediately before and after RF. Consequently, FV/FM and ФPSII of agar inoculates were measured repeatedly for 30 d cultivation in 3 d interval. Both the 32 and the 50 mM addition of ribitol caused earlier detection of the parameters (d 16) compared to the control measurements (d 23) as well as reaching the maximum values of the chlorophyll fluorescence parameters earlier (d 23 in ribitol-treated samples compared to d 25 in control samples). Heterococcus sp. proved to be a species resistant to rapid freezing. The ability may help the species to survive in harsh Antarctic environments typified by rapid fluctuations in temperature that may bring a rapid freezing of the alga.


Subject(s)
Adaptation, Physiological/physiology , Cell Survival/physiology , Chlorophyll/metabolism , Microalgae/physiology , Antarctic Regions , Cell Survival/drug effects , Cold Temperature , Fluorescence , Freezing , Ribitol/metabolism , Ribitol/pharmacology
18.
PLoS One ; 13(2): e0191837, 2018.
Article in English | MEDLINE | ID: mdl-29401462

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are an abundant class of innate T cells restricted by the MHC I-related molecule MR1. MAIT cells can recognize bacterially-derived metabolic intermediates from the riboflavin pathway presented by MR1 and are postulated to play a role in innate antibacterial immunity through production of cytokines and direct bacterial killing. MR1 tetramers, typically stabilized by the adduct of 5-amino-6-D-ribitylaminouracil (5-A-RU) and methylglyoxal (MeG), are important tools for the study of MAIT cells. A long-standing problem with 5-A-RU is that it is unstable upon storage. Herein we report an efficient synthetic approach to the HCl salt of this ligand, which has improved stability during storage. We also show that synthetic 5-A-RU•HCl produced by this method may be used in protocols for the stimulation of human MAIT cells and production of both human and mouse MR1 tetramers for MAIT cell identification.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Minor Histocompatibility Antigens/metabolism , Ribitol/analogs & derivatives , Uracil/analogs & derivatives , Humans , Immunity, Innate , Ligands , Mucosal-Associated Invariant T Cells/immunology , Ribitol/chemical synthesis , Ribitol/metabolism , Uracil/chemical synthesis , Uracil/metabolism
19.
Biochem Biophys Res Commun ; 497(4): 1025-1030, 2018 03 18.
Article in English | MEDLINE | ID: mdl-29477842

ABSTRACT

Dystroglycanopathies are a group of muscular dystrophies that are caused by abnormal glycosylation of dystroglycan; currently 18 causative genes are known. Functions of the dystroglycanopathy genes fukutin, fukutin-related protein (FKRP), and transmembrane protein 5 (TMEM5) were most recently identified; fukutin and FKRP are ribitol-phosphate transferases and TMEM5 is a ribitol xylosyltransferase. In this study, we show that fukutin, FKRP, and TMEM5 form a complex while maintaining each of their enzyme activities. Immunoprecipitation and immunofluorescence experiments demonstrated protein interactions between these 3 proteins. A protein complex consisting of endogenous fukutin and FKRP, and exogenously expressed TMEM5 exerts activities of each enzyme. Our data showed for the first time that endogenous fukutin and FKRP enzyme activities coexist with TMEM5 enzyme activity, and suggest the possibility that formation of this enzyme complex may contribute to specific and prompt biosynthesis of glycans that are required for dystroglycan function.


Subject(s)
Membrane Proteins/metabolism , Muscular Dystrophies/metabolism , Proteins/metabolism , Dystroglycans , HEK293 Cells , Humans , Multiprotein Complexes , Pentosyltransferases , Polysaccharides/biosynthesis , Ribitol/metabolism
20.
J Biol Chem ; 292(43): 17832-17844, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28912268

ABSTRACT

Wall teichoic acids (WTAs) are the most abundant glycopolymers found on the cell wall of many Gram-positive bacteria, whose diverse surface structures play key roles in multiple biological processes. Despite recent technological advances in glycan analysis, structural elucidation of WTAs remains challenging due to their complex nature. Here, we employed a combination of ultra-performance liquid chromatography-coupled electrospray ionization tandem-MS/MS and NMR to determine the structural complexity of WTAs from Listeria species. We unveiled more than 10 different types of WTA polymers that vary in their linkage and repeating units. Disparity in GlcNAc to ribitol connectivity, as well as variable O-acetylation and glycosylation of GlcNAc contribute to the structural diversity of WTAs. Notably, SPR analysis indicated that constitution of WTA determines the recognition by bacteriophage endolysins. Collectively, these findings provide detailed insight into Listeria cell wall-associated carbohydrates, and will guide further studies on the structure-function relationship of WTAs.


Subject(s)
Cell Wall/chemistry , Cell Wall/metabolism , Listeria/metabolism , Teichoic Acids/chemistry , Teichoic Acids/metabolism , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Ribitol/chemistry , Ribitol/metabolism , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL