Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(23)2022 Dec 02.
Article in English | MEDLINE | ID: mdl-36499496

ABSTRACT

Phenolic acids are known flavonoid metabolites, which typically undergo bioconjugation during phase II of biotransformation, forming sulfates, along with other conjugates. Sulfated derivatives of phenolic acids can be synthesized by two approaches: chemoenzymatically by 3'-phosphoadenosine-5'-phosphosulfate (PAPS)-dependent sulfotransferases or PAPS-independent aryl sulfotransferases such as those from Desulfitobacterium hafniense, or chemically using SO3 complexes. Both approaches were tested with six selected phenolic acids (2-hydroxyphenylacetic acid (2-HPA), 3-hydroxyphenylacetic acid (3-HPA), 4-hydroxyphenylacetic acid (4-HPA), 3,4-dihydroxyphenylacetic acid (DHPA), 3-(4-hydroxyphenyl)propionic acid (4-HPP), and 3,4-dihydroxyphenylpropionic acid (DHPP)) to create a library of sulfated metabolites of phenolic acids. The sulfates of 3-HPA, 4-HPA, 4-HPP, DHPA, and DHPP were all obtained by the methods of chemical synthesis. In contrast, the enzymatic sulfation of monohydroxyphenolic acids failed probably due to enzyme inhibition, whereas the same reaction was successful for dihydroxyphenolic acids (DHPA and DHPP). Special attention was also paid to the counterions of the sulfates, a topic often poorly reported in synthetic works. The products obtained will serve as authentic analytical standards in metabolic studies and to determine their biological activity.


Subject(s)
Phosphoadenosine Phosphosulfate , Sulfotransferases , Phosphoadenosine Phosphosulfate/chemistry , Phosphoadenosine Phosphosulfate/metabolism , Sulfotransferases/metabolism , Sulfates/metabolism , Hydroxybenzoates
2.
Biochem Biophys Res Commun ; 586: 1-7, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34818583

ABSTRACT

Sulfation is an essential modification on biomolecules in living cells, and 3'-Phosphoadenosine-5'-phosphosulfate (PAPS) is its unique and universal sulfate donor. Human PAPS synthases (PAPSS1 and 2) are the only enzymes that catalyze PAPS production from inorganic sulfate. Unexpectedly, PAPSS1 and PAPSS2 do not functional complement with each other, and abnormal function of PAPSS2 but not PAPSS1 leads to numerous human diseases including bone development diseases, hormone disorder and cancers. Here, we reported the crystal structures of ATP-sulfurylase domain of human PAPSS2 (ATPS2) and ATPS2 in complex with is product 5'-phosphosulfate (APS). We demonstrated that ATPS2 recognizes the substrates by using family conserved residues located on the HXXH and PP motifs, and achieves substrate binding and releasing by employing a non-conserved phenylalanine (Phe550) through a never observed flipping mechanism. Our discovery provides additional information to better understand the biological function of PAPSS2 especially in tumorigenesis, and may facilitate the drug discovery against this enzyme.


Subject(s)
Adenosine Triphosphate/chemistry , Multienzyme Complexes/chemistry , Neoplasm Proteins/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Sulfate Adenylyltransferase/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Sulfate Adenylyltransferase/genetics , Sulfate Adenylyltransferase/metabolism , Thermodynamics
3.
Org Biomol Chem ; 20(3): 596-605, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34951618

ABSTRACT

Sulfotransferases constitute a ubiquitous class of enzymes which are poorly understood due to the lack of a convenient tool for screening their activity. These enzymes use the anion PAPS (adenosine-3'-phosphate-5'-phosphosulfate) as a donor for a broad range of acceptor substrates, including carbohydrates, producing sulfated compounds and PAP (adenosine-3',5'-diphosphate) as a side product. We present a europium(III)-based probe that binds reversibly to both PAPS and PAP, producing a larger luminescence enhancement with the latter anion. We exploit this greater emission enhancement with PAP to demonstrate the first direct real-time assay of a heparan sulfate sulfotransferase using a multi-well plate format. The selective response of our probe towards PAP over structurally similar nucleoside phosphate anions, and over other anions, is investigated and discussed. This work opens the possibility of investigating more fully the roles played by this enzyme class in health and disease, including operationally simple inhibitor screening.


Subject(s)
Coordination Complexes/metabolism , Europium/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Sulfotransferases/metabolism , Anions/chemistry , Anions/metabolism , Cations/chemistry , Cations/metabolism , Coordination Complexes/chemistry , Europium/chemistry , Molecular Structure , Phosphoadenosine Phosphosulfate/chemistry , Sulfotransferases/chemistry , Time Factors
4.
Nat Prod Rep ; 37(10): 1316-1333, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32582886

ABSTRACT

Covering: up to the beginning of 2020Enzymes depending on cofactors are essential in many biosynthetic pathways of natural products. They are often involved in key steps: catalytic conversions that are difficult to achieve purely with synthetic organic chemistry. Hence, cofactor-dependent enzymes have great potential for biocatalysis, on the condition that a corresponding cofactor regeneration system is available. For some cofactors, these regeneration systems require multiple steps; such complex enzyme cascades/multi-enzyme systems are (still) challenging for in vitro biocatalysis. Further, artificial cofactor analogues have been synthesised that are more stable, show an altered reaction range, or act as inhibitors. The development of bio-orthogonal systems that can be used for the production of modified natural products in vivo is an ongoing challenge. In light of the recent progress in this field, this review aims to provide an overview of general strategies involving enzyme cofactors, cofactor analogues, and regeneration systems; highlighting the current possibilities for application of enzymes using some of the most common cofactors.


Subject(s)
Coenzymes/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Catalysis , Coenzyme A/chemistry , Coenzyme A/metabolism , Coenzymes/chemical synthesis , NADP/chemistry , NADP/metabolism , Nucleosides/metabolism , Phosphoadenosine Phosphosulfate/chemistry , Phosphoadenosine Phosphosulfate/metabolism , Phosphorylation
5.
Biomolecules ; 10(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31935947

ABSTRACT

Sulfation is a common modification of extracellular glycans, tyrosine residues on proteins, and steroid hormones, and is important in a wide variety of signaling pathways. We investigated the role of sulfation on endogenous oxidative stress, such as glutamate-induced oxytosis and erastin-induced ferroptosis, using mouse hippocampal HT22 cells. Sodium chlorate competitively inhibits the formation of 3'-phosphoadenosine 5'-phosphosulfate, the high energy sulfate donor in cellular sulfation reactions. The treatment of HT22 cells with sodium chlorate decreased sulfation of heparan sulfate proteoglycans and chondroitin sulfate proteoglycans. Sodium chlorate and ß-d-xyloside, which prevents proteoglycan glycosaminoglycan chain attachment, exacerbated both glutamate- and erastin-induced cell death, suggesting that extracellular matrix influenced oxytosis and ferroptosis. Moreover, sodium chlorate enhanced the generation of reactive oxygen species and influx of extracellular Ca2+ in the process of oxytosis and ferroptosis. Interestingly, sodium chlorate did not affect antioxidant glutathione levels. Western blot analysis revealed that sodium chlorate enhanced erastin-induced c-Jun N-terminal kinase phosphorylation, which is preferentially activated by cell stress-inducing signals. Collectively, our findings indicate that sulfation is an important modification for neuroprotection against oxytosis and ferroptosis in neuronal hippocampal cells.


Subject(s)
Ferroptosis/physiology , Regulated Cell Death/physiology , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cell Line , Chlorates/pharmacology , Ferroptosis/drug effects , Glutamic Acid/metabolism , Glutathione/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphoadenosine Phosphosulfate/chemistry , Proteoglycans/chemistry , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Somatomedins/metabolism
6.
J Mol Model ; 21(4): 72, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25750022

ABSTRACT

Human cytosolic sulfotransferases (hSULTs) are important phase II metabolic enzymes. They catalyze transfer of the sulfuryl-group (-SO3) from 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to the hydroxyl or primary amine moieties of a large number of endogenous and xenobiotic substrates. Broad selectivity and specificity of binding and activity within the sulfortransferases family could be detected by thermal denaturation assays, which have been made more and more suitable for high throughput screening based on recent technical advances. Here molecular dynamics simulations were used to explore the effect of the cofactor (PAPS) and substrate (LCA) on the thermal stability of the enzyme. It was found that the apo-enzyme unfolded fastest upon heating. The holo-enzyme with bound substrate LCA unfolded slowest. This thermo-denaturation order is consistent with that observed in experiments. Further it was found that the cofactor and substrate will pronouncedly increase the thermal stability of the active pocket regions that interact directly with the ligands. In addition, cofactor and substrate show noticeable synergy effect on the thermal stability of the enzyme.


Subject(s)
Models, Molecular , Molecular Dynamics Simulation , Sulfotransferases/chemistry , Binding Sites , Catalysis , Enzyme Stability , Humans , Ligands , Phosphoadenosine Phosphosulfate/chemistry , Protein Conformation , Substrate Specificity , Sulfotransferases/metabolism , Temperature
7.
Methods Mol Biol ; 1229: 37-42, 2015.
Article in English | MEDLINE | ID: mdl-25325942

ABSTRACT

The biological activity of glycosaminoglycans (GAGs) depends greatly on the sulfation pattern present within the GAG chain. Chemical biology of GAGs can be further advanced by preparation of sulfur-isotope-enriched sulfated GAGs. 3'-Phosphoadenosine-5'-phosphosulfate (PAPS) serves as a universal sulfate donor in the sulfation of GAGs by sulfotransferases. Therefore, synthesis of PAPS carrying sulfur isotopes is critical in the preparation of labeled GAGs for biochemical studies. Here we describe a robust in vitro enzymatic synthesis of sulfur isotope-enriched PAPS which allows for heavy- or radio-isotope labeling of GAG chains.


Subject(s)
Biochemistry/methods , Glycosaminoglycans/metabolism , Phosphoadenosine Phosphosulfate/chemical synthesis , Sulfates/metabolism , Chromatography, Thin Layer , Mass Spectrometry , Phosphoadenosine Phosphosulfate/chemistry , Reproducibility of Results , Sulfur Radioisotopes
8.
Glycobiology ; 25(1): 30-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25138304

ABSTRACT

Proteoglycan (PG) sulfation depends on activated nucleotide sulfate, 3'-phosphoadenosine-5'-phosphosulfate (PAPS). Transporters in the Golgi membrane translocate PAPS from the cytoplasm into the organelle lumen where PG sulfation occurs. Silencing of PAPS transporter (PAPST) 1 in epithelial MDCK cells reduced PAPS uptake into Golgi vesicles. Surprisingly, at the same time sulfation of heparan sulfate (HS) was stimulated. The effect was pathway specific in polarized epithelial cells. Basolaterally secreted proteoglycans (PGs) displayed an altered HS sulfation pattern and increased growth factor binding capacity. In contrast, the sulfation pattern of apically secreted PGs was unchanged while the secretion was reduced. Regulation of PAPST1 allows epithelial cells to prioritize between PG sulfation in the apical and basolateral secretory routes at the level of the Golgi apparatus. This provides sulfation patterns that ensure PG functions at the extracellular level, such as growth factor binding.


Subject(s)
Chondroitin Sulfates/metabolism , Golgi Apparatus/metabolism , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/metabolism , Membrane Transport Proteins/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Animals , Biological Transport , Cell Polarity , Chondroitin Sulfates/chemistry , Dogs , Gene Expression Regulation , Heparan Sulfate Proteoglycans/chemistry , Heparitin Sulfate/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Madin Darby Canine Kidney Cells , Membrane Transport Proteins/genetics , Phosphoadenosine Phosphosulfate/chemistry , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
9.
Biochemistry ; 53(44): 6893-900, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25314023

ABSTRACT

Human cytosolic sulfotransferases (SULTs) regulate the activities of thousands of small molecules-metabolites, drugs, and other xenobiotics-via the transfer of the sulfuryl moiety (-SO3) from 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to the hydroxyls and primary amines of acceptors. SULT1A1 is the most abundant SULT in liver and has the broadest substrate spectrum of any SULT. Here we present the discovery of a new form of SULT1A1 allosteric regulation that modulates the catalytic efficiency of the enzyme over a 130-fold dynamic range. The molecular basis of the regulation is explored in detail and is shown to be rooted in an energetic coupling between the active-site caps of adjacent subunits in the SULT1A1 dimer. The first nucleotide to bind causes closure of the cap to which it is bound and at the same time stabilizes the cap in the adjacent subunit in the open position. Binding of the second nucleotide causes both caps to open. Cap closure sterically controls active-site access of the nucleotide and acceptor; consequently, the structural changes in the cap that occur as a function of nucleotide occupancy lead to changes in the substrate affinities and turnover of the enzyme. PAPS levels in tissues from a variety of organs suggest that the catalytic efficiency of the enzyme varies across tissues over the full 130-fold range and that efficiency is greatest in those tissues that experience the greatest xenobiotic "load".


Subject(s)
Arylsulfotransferase/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Allosteric Regulation , Enzyme Activation , Humans , Kinetics , Protein Binding
10.
J Biol Chem ; 289(38): 26474-26480, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25056952

ABSTRACT

Human cytosolic sulfotransferases (SULTs) regulate the activities of thousands of signaling small molecules via transfer of the sulfuryl moiety (-SO3) from 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to the hydroxyls and primary amines of acceptors. Sulfonation controls the affinities of ligands for their targets, and thereby regulates numerous receptors, which, in turn, regulate complex cellular responses. Despite their biological and medical relevance, basic SULT mechanism issues remain unresolved. To settle these issues, and to create an in-depth model of SULT catalysis, the complete kinetic mechanism of a representative member of the human SULT family, SULT2A1, was determined. The mechanism is composed of eight enzyme forms that interconvert via 22 rate constants, each of which was determined independently. The result is a complete quantitative description of the mechanism that accurately predicts complex enzymatic behavior. This is the first description of a SULT mechanism at this resolution, and it reveals numerous principles of SULT catalysis and resolves previously ambiguous issues. The structures and catalytic behaviors SULTs are highly conserved; hence, the mechanism presented here should prove paradigmatic for the family.


Subject(s)
Sulfotransferases/chemistry , Biocatalysis , Dehydroepiandrosterone/chemistry , Humans , Kinetics , Models, Chemical , Phosphoadenosine Phosphosulfate/chemistry , Protein Binding , Sulfotransferases/antagonists & inhibitors
11.
PLoS One ; 8(8): e70880, 2013.
Article in English | MEDLINE | ID: mdl-23940657

ABSTRACT

Sulfation patterns along glycosaminoglycan (GAG) chains dictate their functional role. The N-deacetylase N-sulfotransferase family (NDST) catalyzes the initial downstream modification of heparan sulfate and heparin chains by removing acetyl groups from subsets of N-acetylglucosamine units and, subsequently, sulfating the residual free amino groups. These enzymes transfer the sulfuryl group from 3'-phosphoadenosine-5'-phosphosulfate (PAPS), yielding sulfated sugar chains and 3'-phosphoadenosine-5'-phosphate (PAP). For the N-sulfotransferase domain of NDST1, Lys833 has been implicated to play a role in holding the substrate glycan moiety close to the PAPS cofactor. Additionally, Lys833 together with His716 interact with the sulfonate group, stabilizing the transition state. Such a role seems to be shared by Lys614 through donation of a proton to the bridging oxygen of the cofactor, thereby acting as a catalytic acid. However, the relevance of these boundary residues at the hydrophobic cleft is still unclear. Moreover, whether Lys833, His716 and Lys614 play a role in both glycan recognition and glycan sulfation remains elusive. In this study we evaluate the contribution of NDST mutants (Lys833, His716 and Lys614) to dynamical effects during sulfate transfer using comprehensive combined docking and essential dynamics. In addition, the binding location of the glycan moiety, PAPS and PAP within the active site of NDST1 throughout the sulfate transfer were determined by intermediate state analysis. Furthermore, NDST1 mutants unveiled Lys833 as vital for both the glycan binding and subsequent N-sulfotransferase activity of NDST1.


Subject(s)
Molecular Dynamics Simulation , Sulfotransferases/chemistry , Amino Acid Substitution , Catalytic Domain , Disaccharides/chemistry , Heparitin Sulfate/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mutant Proteins/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Protein Binding , Sulfotransferases/genetics
12.
Mol Cell Proteomics ; 12(4): 979-90, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23429520

ABSTRACT

The highly sulfated glycosaminoglycan oligosaccharides derived from heparin and heparan sulfate have been a highly intractable class of molecules to analyze by tandem mass spectrometry. Under the many methods of ion activation, this class of molecules generally exhibits SO3 loss as the most significant fragmentation pathway, interfering with the assignment of the location of sulfo groups in glycosaminoglycan chains. We report here a method that stabilizes sulfo groups and facilitates the complete structural analysis of densely sulfated (two or more sulfo groups per disaccharide repeat unit) heparin and heparan sulfate oligomers. This is achieved by complete removal of all ionizable protons, either by charging during electrospray ionization or by Na(+)/H(+) exchange. The addition of millimolar levels of NaOH to the sample solution facilitates the production of precursor ions that meet this criterion. This approach is found to work for a variety of heparin sulfate oligosaccharides derived from natural sources or produced by chemoenzymatic synthesis, with up to 12 saccharide subunits and up to 11 sulfo groups.


Subject(s)
Heparin/chemistry , Heparitin Sulfate/chemistry , Sulfotransferases/chemistry , Tandem Mass Spectrometry , Animals , Biocatalysis , Carbohydrate Conformation , Carbohydrate Sequence , Heparin/biosynthesis , Heparitin Sulfate/biosynthesis , Molecular Sequence Data , Oligosaccharides/biosynthesis , Oligosaccharides/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Spectrometry, Mass, Electrospray Ionization , Sulfuric Acid Esters/chemistry , Sus scrofa
13.
Biochemistry ; 51(28): 5674-83, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22703301

ABSTRACT

Human SULT2A1 is one of two predominant sulfotransferases in liver and catalyzes transfer of the sulfuryl moiety (-SO(3)) from activated sulfate (PAPS, 3'-phosphoadenosine 5-phosphosulfate) to hundreds of acceptors (metabolites and xenobiotics). Sulfation recodes the biologic activity of acceptors by altering their receptor interactions. The molecular basis on which these enzymes select and sulfonate specific acceptors from complex mixtures of competitors in vivo is a long-standing issue in the SULT field. Raloxifene, a synthetic steroid used in the prevention of osteoporosis, and dehydroepiandrosterone (DHEA), a ubiquitous steroid precusor, are reported to be sulfated efficiently by SULT2A1 in vitro, yet unlike DHEA, raloxifene is not sulfated in vivo. This selectivity was explored in initial rate and equilibrium binding studies that demonstrate pronounced binding antisynergy (21-fold) between PAPS and raloxifene, but not DHEA. Analysis of crystal structures suggests that PAP binding restricts access to the acceptor-binding pocket by restructuring a nine-residue segment of the pocket edge that constricts the active site opening, or "pore", that sieves substrates on the basis of their geometries. In silico docking predicts that raloxifene, which is considerably larger than DHEA, can bind only to the unliganded (open) enzyme, whereas DHEA binds both the open and closed forms. The predictions of these structures with regard to substrate binding are tested using equilibrium and pre-steady-state ligand binding studies, and the results confirm that a nucleotide-driven isomerization controls access to the acceptor-binding pocket and plays an important role in substrate selection by SULT2A1 and possibly other sulfotransferases.


Subject(s)
Bone Density Conservation Agents/chemistry , Dehydroepiandrosterone/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Raloxifene Hydrochloride/chemistry , Sulfotransferases/chemistry , Computer Simulation , Kinetics , Molecular Docking Simulation , Protein Binding , Recombinant Proteins/chemistry , Substrate Specificity
14.
Bioorg Med Chem Lett ; 22(11): 3661-4, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22572581

ABSTRACT

We describe an efficient and scalable procedure for the chemical synthesis of nucleoside 5'-phosphosulfates (NPS) from nucleoside 5'-phosphorimidazolides and sulfate bis(tributylammonium) salt. Using this method we obtained various NPS with yields ranging from 70-90%, including adenosine 5'-phosphosulfate (APS) and 2',3'-cyclic precursor of 3'-phosphoadenosine 5'-phosphosulfate (PAPS), which are the key intermediates in the assimilation and metabolism of sulfur in all living organisms.


Subject(s)
Adenosine Phosphosulfate/chemistry , Nucleosides/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Adenosine Phosphosulfate/chemical synthesis , Eukaryotic Initiation Factor-4E/metabolism , Protein Binding
15.
Expert Opin Drug Metab Toxicol ; 8(6): 635-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22512672

ABSTRACT

INTRODUCTION: Cytosolic sulfotransferases (SULTs) are the enzymes that catalyze the sulfonation reaction, an important metabolic pathway for numerous endogenous and exogenous compounds. Human SULTs exhibit complex patterns of broad, differential and overlapping substrate selectivity. Moreover, these enzymes often display substrate inhibition kinetics (i.e., inhibition of the enzyme activity at high substrate concentrations). AREAS COVERED: At present, the crystal structures for 12 human SULTs (i.e., SULT1A1, 1A2, 1A3, 1B1, 1C1, 1C2, 1C3, 1E1, 2A1, 2B1a, 2B1b and 4A1) are available, many of which are in complex with a substrate. This review describes the similarities and differences in these structures (particularly the active-site structures) of SULT enzymes. The authors also discuss the structural basis for understanding the catalytic mechanism, the substrate inhibition mechanisms, the cofactor (3'-phosphoadenosine 5'-phosphosulfate or PAPS) binding and the substrate recognition. EXPERT OPINION: Correlations of the structural features (including conformational flexibility) in the active sites with the substrate profiles of several SULTs have been well established. One is encouraged to closely integrate in silico approaches with the structural knowledge of the active sites for development of a rationalized and accurate tool that is able to predict metabolism of SULTs toward chemicals and drug candidates.


Subject(s)
Cytosol/enzymology , Sulfotransferases/chemistry , Amino Acid Sequence , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Humans , Metabolic Detoxication, Phase II , Models, Molecular , Molecular Sequence Data , Phosphoadenosine Phosphosulfate/chemistry , Phosphoadenosine Phosphosulfate/metabolism , Protein Conformation , Structure-Activity Relationship , Substrate Specificity , Sulfotransferases/metabolism
17.
Glycobiology ; 21(6): 771-80, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21224284

ABSTRACT

Heparan sulfate (HS) belongs to a major class of glycans that perform central physiological functions. Heparin is a specialized form of HS and is a clinically used anticoagulant drug. Heparin is a natural product isolated from pig intestine. There is a strong demand to replace natural heparin with a synthetic counterpart. Although a chemoenzymatic approach has been employed to prepare synthetic heparin, the scale of the synthesis is limited by the availability of sulfotransferases and the cofactor, 3'-phosphoadenosine-5'-phosphosulfate (PAPS). Here, we present a novel method to produce secreted forms of sulfotransferases in the yeast cells, Kluyveromyces lactis. Five sulfotransferases including N-sulfotransferase, 2-O-sulfotransferase, 3-O-sulfotransferase 1 and 6-O-sulfotransferases 1 and 3 were expressed using this method. Unlike bacterial-expressed sulfotransferases, the yeast proteins can be directly used to modify polysaccharides without laborious purification. The yeast-expressed sulfotransferases also tend to have higher specific activity and thermostability. Furthermore, we demonstrated the possibility for the gram-scale synthesis of PAPS from adenosine 5'-triphosphate at only 1/5000th of the price purchased from a commercial source. Our results pave the way to conduct the enzymatic synthesis of heparin in large quantities.


Subject(s)
Kluyveromyces/enzymology , Phosphoadenosine Phosphosulfate/biosynthesis , Sulfotransferases/biosynthesis , Carbohydrate Conformation , Gene Expression , Phosphoadenosine Phosphosulfate/chemistry , Phosphoadenosine Phosphosulfate/isolation & purification , Polysaccharides/biosynthesis , Polysaccharides/chemistry , Sulfotransferases/isolation & purification , Sulfotransferases/metabolism
18.
Anal Chem ; 82(13): 5887-91, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20521766

ABSTRACT

We propose a novel strategy for determining the elemental composition of organic compounds using the peak ratio of isotopic fine structure observed by high-magnetic field Fourier transform ion cyclotron resonance mass spectrometry (FT-ICR-MS). Using 3'-phosphoadenosine 5'-phosphosulfate and CTU guanamine as standard organic compounds, isotopic peaks derived from (15)N-, (34)S-, and (18)O-substituted forms were separated from (13)C-substituted species. Furthermore, these isotopic peaks were quantitatively detected and closely matched the natural abundance of each element. These data successfully led us to determine the one elemental composition in a standard independent manner. The approach should be particularly amenable to the metabolomics research field.


Subject(s)
Fourier Analysis , Ions/chemistry , Mass Spectrometry/methods , Organic Chemicals/chemistry , Carbon Isotopes/chemistry , Metabolomics/methods , Nitrogen Isotopes/chemistry , Oxygen Isotopes/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Sulfur Isotopes/chemistry
19.
Biochem Biophys Res Commun ; 396(2): 429-34, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20417180

ABSTRACT

The cytosolic sulfotransferases (SULTs) in vertebrates catalyze the sulfonation of endogenous thyroid/steroid hormones and catecholamine neurotransmitters, as well as a variety of xenobiotics, using 3'-phosphoadenosine 5'-phosphosulfate (PAPS) as the sulfonate donor. In this study, we determined the structures of SULT1A2 and an allozyme of SULT1A1, SULT1A1 *3, bound with 3'-phosphoadenosine 5'-phosphate (PAP), at 2.4 and 2.3A resolution, respectively. The conformational differences between the two structures revealed a plastic substrate-binding pocket with two channels and a switch-like substrate selectivity residue Phe247, providing clearly a structural basis for the substrate inhibition. In SULT1A2, Tyr149 extends approximately 2.1A further to the inside of the substrate-binding pocket, compared with the corresponding His149 residue in SULT1A1 *3. Site-directed mutagenesis study showed that, compared with the wild-type SULT1A2, mutant Tyr149Phe SULT1A2 exhibited a 40 times higher K(m) and two times lower V(max) with p-nitrophenol as substrate. These latter data imply a significant role of Tyr149 in the catalytic mechanism of SULT1A2.


Subject(s)
Arylsulfotransferase/antagonists & inhibitors , Arylsulfotransferase/chemistry , Arylsulfotransferase/genetics , Catalysis , Crystallography, X-Ray , Humans , Mutagenesis, Site-Directed , Mutation , Nitrophenols/chemistry , Phosphoadenosine Phosphosulfate/chemistry , Protein Conformation , Substrate Specificity , Tyrosine/chemistry , Tyrosine/genetics
20.
FEBS Lett ; 583(18): 3091-4, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19695253

ABSTRACT

Mammalian sulfotransferases (STs) utilize exclusively the sulfuryl group donor 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to catalyze the sulfurylation reactions based on a sequential transfer mechanism. In contrast, the commensal intestinal bacterial arylsulfate sulfotransferases (ASSTs) do not use PAPS as the sulfuryl group donor, but instead catalyze sulfuryl transfer from phenolic sulfate to a phenol via a Ping-Pong mechanism. Interestingly, structural comparison revealed a similar spatial arrangement of the active site residues as well as the cognate substrates in mouse ST (mSULT1D1) and Escherichia coli CFT073 ASST, despite that their overall structures bear no discernible relationship. These observations suggest that the active sites of PAPS-dependent SULT1D1 and phenolic sulfate-utilizing ASST represent an example of convergent evolution.


Subject(s)
Arylsulfotransferase/chemistry , Catalytic Domain , Phosphoadenosine Phosphosulfate/chemistry , Animals , Arylsulfotransferase/genetics , Catalysis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Evolution, Molecular , Mice , Phosphoadenosine Phosphosulfate/genetics , Protein Conformation , Sulfotransferases
SELECTION OF CITATIONS
SEARCH DETAIL