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










Publication year range
1.
J Biol Chem ; 298(11): 102505, 2022 11.
Article in English | MEDLINE | ID: mdl-36126773

ABSTRACT

MutS homologs (MSHs) are highly conserved core components of DNA mismatch repair. Mismatch recognition provokes ATP-binding by MSH proteins that drives a conformational transition from a short-lived lesion-searching clamp to an extremely stable sliding clamp on the DNA. Here, we have expanded on previous bulk biochemical studies to examine the stability, lifetime, and kinetics of bacterial and human MSH sliding clamps on mismatched DNA using surface plasmon resonance and single-molecule analysis of fluorescently labeled proteins. We found that ATP-bound MSH complexes bound to blocked-end or very long mismatched DNAs were extremely stable over a range of ionic conditions. These observations underpinned the development of a high-throughput Förster resonance energy transfer system that specifically detects the formation of MSH sliding clamps on mismatched DNA. The Förster resonance energy transfer system is capable of distinguishing between HsMSH2-HsMSH3 and HsMSH2-HsMSH6 and appears suitable for chemical inhibitor screens. Taken together, our results provide additional insight into MSH sliding clamps as well as methods to distinguish their functions in mismatch repair.


Subject(s)
Escherichia coli Proteins , MutS DNA Mismatch-Binding Protein , Humans , Adenosine Triphosphate/metabolism , Base Pair Mismatch , DNA/metabolism , DNA Mismatch Repair , Escherichia coli Proteins/metabolism , MutS DNA Mismatch-Binding Protein/genetics , MutS DNA Mismatch-Binding Protein/metabolism , MutS Homolog 2 Protein/genetics , MutS Homolog 2 Protein/metabolism , MutS Proteins/genetics , Protein Binding
3.
Nat Chem Biol ; 18(8): 841-849, 2022 08.
Article in English | MEDLINE | ID: mdl-35710619

ABSTRACT

Sulfated glycans are ubiquitous nutrient sources for microbial communities that have coevolved with eukaryotic hosts. Bacteria metabolize sulfated glycans by deploying carbohydrate sulfatases that remove sulfate esters. Despite the biological importance of sulfatases, the mechanisms underlying their ability to recognize their glycan substrate remain poorly understood. Here, we use structural biology to determine how sulfatases from the human gut microbiota recognize sulfated glycans. We reveal seven new carbohydrate sulfatase structures spanning four S1 sulfatase subfamilies. Structures of S1_16 and S1_46 represent novel structures of these subfamilies. Structures of S1_11 and S1_15 demonstrate how non-conserved regions of the protein drive specificity toward related but distinct glycan targets. Collectively, these data reveal that carbohydrate sulfatases are highly selective for the glycan component of their substrate. These data provide new approaches for probing sulfated glycan metabolism while revealing the roles carbohydrate sulfatases play in host glycan catabolism.


Subject(s)
Gastrointestinal Microbiome , Sulfatases , Bacteria/metabolism , Humans , Polysaccharides/chemistry , Sulfatases/chemistry , Sulfates/chemistry
4.
Nature ; 598(7880): 332-337, 2021 10.
Article in English | MEDLINE | ID: mdl-34616040

ABSTRACT

Humans have co-evolved with a dense community of microbial symbionts that inhabit the lower intestine. In the colon, secreted mucus creates a barrier that separates these microorganisms from the intestinal epithelium1. Some gut bacteria are able to utilize mucin glycoproteins, the main mucus component, as a nutrient source. However, it remains unclear which bacterial enzymes initiate degradation of the complex O-glycans found in mucins. In the distal colon, these glycans are heavily sulfated, but specific sulfatases that are active on colonic mucins have not been identified. Here we show that sulfatases are essential to the utilization of distal colonic mucin O-glycans by the human gut symbiont Bacteroides thetaiotaomicron. We characterized the activity of 12 different sulfatases produced by this species, showing that they are collectively active on all known sulfate linkages in O-glycans. Crystal structures of three enzymes provide mechanistic insight into the molecular basis of substrate specificity. Unexpectedly, we found that a single sulfatase is essential for utilization of sulfated O-glycans in vitro and also has a major role in vivo. Our results provide insight into the mechanisms of mucin degradation by a prominent group of gut bacteria, an important process for both normal microbial gut colonization2 and diseases such as inflammatory bowel disease3.


Subject(s)
Bacteroides/enzymology , Colon/metabolism , Colon/microbiology , Gastrointestinal Microbiome , Mucins/metabolism , Sulfatases/metabolism , Acetylgalactosamine/chemistry , Acetylgalactosamine/metabolism , Animals , Colon/chemistry , Crystallography, X-Ray , Female , Galactose/metabolism , Humans , Male , Mice , Models, Molecular , Substrate Specificity , Sulfatases/chemistry
5.
Molecules ; 26(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34500644

ABSTRACT

The linear anionic class of polysaccharides, glycosaminoglycans (GAGs), are critical throughout the animal kingdom for developmental processes and the maintenance of healthy tissues. They are also of interest as a means of influencing biochemical processes. One member of the GAG family, heparin, is exploited globally as a major anticoagulant pharmaceutical and there is a growing interest in the potential of other GAGs for diverse applications ranging from skin care to the treatment of neurodegenerative conditions, and from the treatment and prevention of microbial infection to biotechnology. To realize the potential of GAGs, however, it is necessary to develop effective tools that are able to exploit the chemical manipulations to which GAGs are susceptible. Here, the current knowledge concerning the chemical modification of GAGs, one of the principal approaches for the study of the structure-function relationships in these molecules, is reviewed. Some additional methods that were applied successfully to the analysis and/or processing of other carbohydrates, but which could be suitable in GAG chemistry, are also discussed.


Subject(s)
Glycosaminoglycans/chemistry , Polysaccharides/chemistry , Animals , Anticoagulants/chemistry , Heparin/chemistry , Humans , Structure-Activity Relationship
7.
Biochem J ; 478(4): 735-748, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33480417

ABSTRACT

Sulfated carbohydrate metabolism is a fundamental process, which occurs in all domains of life. Carbohydrate sulfatases are enzymes that remove sulfate groups from carbohydrates and are essential to the depolymerisation of complex polysaccharides. Despite their biological importance, carbohydrate sulfatases are poorly studied and challenges remain in accurately assessing the enzymatic activity, specificity and kinetic parameters. Most notably, the separation of desulfated products from sulfated substrates is currently a time-consuming process. In this paper, we describe the development of rapid capillary electrophoresis coupled to substrate fluorescence detection as a high-throughput and facile means of analysing carbohydrate sulfatase activity. The approach has utility for the determination of both kinetic and inhibition parameters and is based on existing microfluidic technology coupled to a new synthetic fluorescent 6S-GlcNAc carbohydrate substrate. Furthermore, we compare this technique, in terms of both time and resources, to high-performance anion exchange chromatography and NMR-based methods, which are the two current 'gold standards' for enzymatic carbohydrate sulfation analysis. Our study clearly demonstrates the advantages of mobility shift assays for the quantification of near real-time carbohydrate desulfation by purified sulfatases, and will support the search for small molecule inhibitors of these disease-associated enzymes.


Subject(s)
Electrophoresis, Capillary/methods , Electrophoretic Mobility Shift Assay/methods , Fluorometry/methods , High-Throughput Screening Assays/methods , Microfluidic Analytical Techniques/methods , Sulfotransferases/analysis , Bacterial Proteins/analysis , Bacterial Proteins/antagonists & inhibitors , Bacteroides thetaiotaomicron/enzymology , Boron Compounds/analysis , Carbohydrate Conformation , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Computer Systems , Fluorescent Dyes/analysis , Glycosaminoglycans/metabolism , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Recombinant Proteins/analysis , Substrate Specificity , Sulfotransferases/antagonists & inhibitors
8.
Carbohydr Res ; 499: 108225, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33353664

ABSTRACT

The formation of ß-glucuronides is a major route by which mammals detoxify and remove breakdown products, such as l-tyrosine, as well as many xenobiotics, from their systems. In humans, dietary l-tyrosine is broken down largely by the action of the anaerobic gut bacterium C. difficile to p-cresol, providing a competitive advantage in the gut microbiota. Ortho- (o-) and meta- (m-), cresols, also present in the environment, may share a common degradative pathway. Relatively little work has been done on cresyl glucuronides. Here, a direct synthesis of o-, m-, and p-cresyl ß-D-glucuronides from methyl 1,2,3,4 tetra-O-acetyl-ß-d-glucuronate and the respective cresol employing trimethylsilyltriflate as promoter is presented. The protected intermediates were hydrolysed using aqueous sodium carbonate to yield the cresyl ß-glucuronides. The toxicities of the o-, m- and p-cresyl ß-D-glucuronides were compared. All three were less toxic to HEK293 cells than their respective cresol precursors: toxicity followed the order o < m < p for Na+ salts and o < p < m for Ca2+ salts. The m-cresyl-glucuronide Ca2+ salt and p-cresyl-glucuronide Na+ salt reduced colony formation by 11% and 9% (v. 30% reduction from the aglycone) respectively, whereas o-cresyl-glucuronide (both Na+ and Ca2+ salts), mildly stimulated HEK293 cell growth.


Subject(s)
Cresols/pharmacology , Glucuronides/pharmacology , Cell Survival/drug effects , Cresols/chemical synthesis , Cresols/chemistry , Dose-Response Relationship, Drug , Glucuronides/chemical synthesis , Glucuronides/chemistry , HEK293 Cells , Humans , Molecular Structure , Stereoisomerism
9.
Nat Commun ; 10(1): 5294, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31757945

ABSTRACT

A shared paradigm of mismatch repair (MMR) across biology depicts extensive exonuclease-driven strand-specific excision that begins at a distant single-stranded DNA (ssDNA) break and proceeds back past the mismatched nucleotides. Historical reconstitution studies concluded that Escherichia coli (Ec) MMR employed EcMutS, EcMutL, EcMutH, EcUvrD, EcSSB and one of four ssDNA exonucleases to accomplish excision. Recent single-molecule images demonstrated that EcMutS and EcMutL formed cascading sliding clamps on a mismatched DNA that together assisted EcMutH in introducing ssDNA breaks at distant newly replicated GATC sites. Here we visualize the complete strand-specific excision process and find that long-lived EcMutL sliding clamps capture EcUvrD helicase near the ssDNA break, significantly increasing its unwinding processivity. EcSSB modulates the EcMutL-EcUvrD unwinding dynamics, which is rarely accompanied by extensive ssDNA exonuclease digestion. Together these observations are consistent with an exonuclease-independent MMR strand excision mechanism that relies on EcMutL-EcUvrD helicase-driven displacement of ssDNA segments between adjacent EcMutH-GATC incisions.


Subject(s)
DNA Breaks, Single-Stranded , DNA Helicases/physiology , DNA Mismatch Repair/physiology , Escherichia coli Proteins/physiology , Escherichia coli/physiology , MutL Proteins/physiology , DNA Helicases/metabolism , DNA Repair/physiology , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Microscopy, Fluorescence , MutL Proteins/metabolism , Single Molecule Imaging
10.
Carbohydr Polym ; 222: 115031, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31320064

ABSTRACT

The detailed structure of a further Chondroitin Sulfate from Litopenaeus vannamei shrimp (sCS) is described. The backbone structure was established by 1H/13C NMR, which identified 3-O-sulfated GlcA, 4-O-sulfated GalNAc, 6-O-sulfated GalNAc, and 4,6-di-O-sulfated GalNAc residues. GlcA is linked to GalNAc 4,6 di S and GlcA 3S is linked to GalNAc 4S, GalNAc 4,6 di-S and GalNAc6S residues. The anticoagulant properties of this sCS were evaluated by activated partial thromboplastin time, anti-IIa, anti-Xa and anti-heparin cofactor II-mediated activities, and sCS failed to stabilise antithrombin in a fluoresence shift assay. The anti-inflammatory effect of sCS was explored using a model of acute peritonitis, followed by leukocyte count and measurement of the cytokines, IL-1ß, IL-6 and TNF-α. The compound showed low clotting effects, but high anti-IIa activity and HCII-mediated thrombin inhibition. Its anti-inflammatory effect was shown by leukocyte recruitment inhibition and a decrease in pro-inflammatory cytokine levels. Although the biological role of sCS remains unknown, its properties indicate that it is suitable for studies of multi-potent molecules obtained from natural sources.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antithrombins/therapeutic use , Chondroitin Sulfates/therapeutic use , Inflammation/drug therapy , Penaeidae/chemistry , Peritonitis/drug therapy , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Antithrombins/chemistry , Antithrombins/isolation & purification , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/isolation & purification , Cytokines/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Molecular Weight , Nitric Oxide/metabolism , Peritonitis/chemically induced , RAW 264.7 Cells , Rats, Wistar
11.
Front Microbiol ; 9: 3107, 2018.
Article in English | MEDLINE | ID: mdl-30619186

ABSTRACT

HIV-1 infection can be successfully controlled with anti-retroviral therapy (ART), but is not cured. A reservoir of cells harboring transcriptionally silent integrated provirus is able to reestablish replicating infection if ART is stopped. Latently HIV-1 infected cells are rare, but may persist for decades. Several novel strategies have been proposed to reduce the latent reservoir, including DNA sequence targeted CRISPR/Cas9 genome editing of the HIV-1 provirus. A significant challenge to genome editing is the sequence diversity of HIV-1 quasispecies present in patients. The high level of quasispecies diversity will require targeting of multiple sites in the viral genome and personalized engineering of a CRISPR/Cas9 regimen. The challenges of CRISPR/Cas9 delivery to the rare latently infected cells and quasispecies sequence diversity suggest that effective genome editing of every provirus is unlikely. However, recent evidence from post-treatment controllers, patients with controlled HIV-1 viral burden following interruption of ART, suggests a correlation between a reduced number of intact proviral sequences and control of the virus. The possibility of reducing the intact proviral sequences in patients by a genome editing technology remains intriguing, but requires significant advances in delivery to infected cells and identification of effective target sites.

SELECTION OF CITATIONS
SEARCH DETAIL