Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 302
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biosci Biotechnol Biochem ; 88(4): 368-380, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38271594

ABSTRACT

Organisms have conversion systems for sulfate ion to take advantage of the chemical features. The use of biologically converted sulfonucleotides varies in an evolutionary manner, with the universal use being that of sulfonate donors. Sulfotransferases have the ability to transfer the sulfonate group of 3'-phosphoadenosine 5'-phosphosulfate to a variety of molecules. Cytosolic sulfotransferases (SULTs) play a role in the metabolism of low-molecular-weight compounds in response to the host organism's living environment. This review will address the diverse functions of the SULT in evolution, including recent findings. In addition to the diversity of vertebrate sulfotransferases, the molecular aspects and recent studies on bacterial and plant sulfotransferases are also addressed.


Subject(s)
Phosphoadenosine Phosphosulfate , Sulfotransferases , Sulfotransferases/chemistry , Cytosol/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Sulfates/metabolism
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.
Drug Metab Dispos ; 50(7): 1027-1041, 2022 07.
Article in English | MEDLINE | ID: mdl-35197313

ABSTRACT

Sulfotransferases are ubiquitous enzymes that transfer a sulfo group from the universal cofactor donor 3'-phosphoadenosine 5'-phosphosulfate to a broad range of acceptor substrates. In humans, the cytosolic sulfotransferases are involved in the sulfation of endogenous compounds such as steroids, neurotransmitters, hormones, and bile acids as well as xenobiotics including drugs, toxins, and environmental chemicals. The Golgi associated membrane-bound sulfotransferases are involved in post-translational modification of macromolecules from glycosaminoglycans to proteins. The sulfation of small molecules can have profound biologic effects on the functionality of the acceptor, including activation, deactivation, or enhanced metabolism and elimination. Sulfation of macromolecules has been shown to regulate a number of physiologic and pathophysiological pathways by enhancing binding affinity to regulatory proteins or binding partners. Over the last 25 years, crystal structures of these enzymes have provided a wealth of information on the mechanisms of this process and the specificity of these enzymes. This review will focus on the general commonalities of the sulfotransferases, from enzyme structure to catalytic mechanism as well as providing examples into how structural information is being used to either design drugs that inhibit sulfotransferases or to modify the enzymes to improve drug synthesis. SIGNIFICANCE STATEMENT: This manuscript honors Dr. Masahiko Negishi's contribution to the understanding of sulfotransferase mechanism, specificity, and roles in biology by analyzing the crystal structures that have been solved over the last 25 years.


Subject(s)
Glycomics , Sulfotransferases , Humans , Inactivation, Metabolic , Phosphoadenosine Phosphosulfate/metabolism , Steroids , Sulfotransferases/metabolism
4.
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
5.
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
6.
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
7.
Appl Microbiol Biotechnol ; 101(20): 7535-7544, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28920175

ABSTRACT

3'-Phosphoadenosine-5'-phosphosulfate (PAPS) is the obligate cosubstrate and source of the sulfonate group in the chemoenzymatic synthesis of heparin, a commonly used anticoagulant drug. Previously, using ATP as the substrate, we had developed a one-pot synthesis to prepare PAPS with 47% ATP conversion efficiency. During the reaction, 47% of ATP was converted into the by-product, ADP. Here, to increase the conversion ratio of ATP to PAPS, an ATP regeneration system was developed to couple with PAPS synthesis. In the ATP regeneration system, the chemical compound, monopotassium phosphoenolpyruvate (PEP-K+), was synthesized and used as the phospho-donor. By using 3-bromopyruvic acid as the starting material, the total yield of PEP-K+ synthesis was over 50% at low cost. Then, the enzyme PykA from Escherichia coli was overexpressed, purified, and used to convert the by-product ADP into ATP. When coupled the ATP regeneration system with PAPS synthesis, the higher ratio of PEP-K+ to ADP was associated with higher ATP conversion efficiency. By using the ATP regeneration system, the conversion ratio of ATP to PAPS was increased to 98% as determined by PAMN-HPLC analysis, and 5Ā g of PAPS was produced in 1Ā L of the reaction mixture. Furthermore, the chemoenzymatic synthesized PAPS was purified and freeze-dried without observed decomposition. However, the powdery PAPS was more unstable than the PAPS sodium salt in aqueous solution at ambient temperature. This developed chemoenzymatic approach of PAPS production will contribute to the synthesis of heparin, in which PAPS is necessary as the individual sulfo-donor.


Subject(s)
Adenosine Triphosphate/metabolism , Escherichia coli Proteins/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Phosphoenolpyruvate/chemical synthesis , Phosphoenolpyruvate/metabolism , Pyruvate Kinase/metabolism
8.
Biochemistry ; 55(29): 4036-46, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27356022

ABSTRACT

The human cytosolic sulfotransferases (SULTs) comprise a 13-member enzyme family that regulates the activities of hundreds, perhaps thousands, of signaling small molecules via regiospecific transfer of the sulfuryl moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and amines of acceptors. Signaling molecules regulated by sulfonation include numerous steroid and thyroid hormones, epinephrine, serotonin, and dopamine. SULT1A1, a major phase II metabolism SULT isoform, is found at a high concentration in liver and has recently been show to harbor two allosteric binding sites, each of which binds a separate and complex class of compounds: the catechins (naturally occurring polyphenols) and nonsteroidal anti-inflammatory drugs. Among catechins, epigallocatechin gallate (EGCG) displays high affinity and specificity for SULT1A1. The allosteric network associated with either site has yet to be defined. Here, using equilibrium binding and pre-steady state studies, the network is shown to involve 14 distinct complexes. ECGG binds both the allosteric site and, relatively weakly, the active site of SULT1A1. It is not a SULT1A1 substrate but is sulfonated by SULT2A1. EGCG binds 17-fold more tightly when the active-site cap of the enzyme is closed by the binding of the nucleotide. When nucleotide is saturating, EGCG binds in two phases. In the first, it binds to the cap-open conformer; in the second, it traps the cap in the closed configuration. Cap closure encapsulates the nucleotide, preventing its release; hence, the EGCG-induced cap stabilization slows nucleotide release, inhibiting turnover. Finally, a comprehensive quantitative model of the network is presented.


Subject(s)
Arylsulfotransferase/chemistry , Arylsulfotransferase/metabolism , Allosteric Regulation , Allosteric Site , Catechin/analogs & derivatives , Catechin/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Ligands , Phosphoadenosine Phosphosulfate/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
9.
Eukaryot Cell ; 14(11): 1144-50, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26385892

ABSTRACT

Entamoeba histolytica, a microaerophilic protozoan parasite, possesses mitosomes. Mitosomes are mitochondrion-related organelles that have largely lost typical mitochondrial functions, such as those involved in the tricarboxylic acid cycle and oxidative phosphorylation. The biological roles of Entamoeba mitosomes have been a long-standing enigma. We previously demonstrated that sulfate activation, which is not generally compartmentalized to mitochondria, is a major function of E. histolytica mitosomes. Sulfate activation cooperates with cytosolic enzymes, i.e., sulfotransferases (SULTs), for the synthesis of sulfolipids, one of which is cholesteryl sulfate. Notably, cholesteryl sulfate plays an important role in encystation, an essential process in the Entamoeba life cycle. These findings identified a biological role for Entamoeba mitosomes; however, they simultaneously raised a new issue concerning how the reactions of the pathway, separated by the mitosomal membranes, cooperate. Here, we demonstrated that the E. histolytica mitochondrial carrier family (EhMCF) has the capacity to exchange 3'-phosphoadenosine 5'-phosphosulfate (PAPS) with ATP. We also confirmed the cytosolic localization of all the E. histolytica SULTs, suggesting that in Entamoeba, PAPS, which is produced through mitosomal sulfate activation, is translocated to the cytosol and becomes a substrate for SULTs. In contrast, ATP, which is produced through cytosolic pathways, is translocated into the mitosomes and is a necessary substrate for sulfate activation. Taking our findings collectively, we suggest that EhMCF functions as a PAPS/ATP antiporter and plays a crucial role in linking the mitosomal sulfate activation pathway to cytosolic SULTs for the production of sulfolipids.


Subject(s)
Adenosine Triphosphate/metabolism , Entamoeba histolytica/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Sulfotransferases/metabolism , Cytoplasm/metabolism , Entamoeba histolytica/genetics , Lipids/biosynthesis , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Sulfotransferases/genetics
10.
Biochemistry ; 54(45): 6830-41, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26512869

ABSTRACT

In many organisms, 3'-phosphoadenosine 5'-phosphate (PAP) is a product of two reactions in the sulfur activation pathway. The sulfurylation of biomolecules, catalyzed by sulfotransferases, uses 3'-phosphoadenosine 5'-phosphosulfate (PAPS) as a sulfate donor, producing the sulfated biomolecule and PAP product. Additionally, the first step in sulfate reduction for many bacteria and fungi reduces the sulfate moiety of PAPS, producing PAP and sulfite, which is subsequently reduced to sulfide. PAP is removed by the phosphatase activity of CysQ, a 3',5'-bisphosphate nucleotidase, yielding AMP and phosphate. Because excess PAP alters the equilibrium of the sulfur pathway and inhibits sulfotransferases, PAP concentrations can affect the levels of sulfur-containing metabolites. Therefore, CysQ, a divalent cation metal-dependent phosphatase, is a major regulator of this pathway. CysQ (Rv2131c) from Mycobacterium tuberculosis (Mtb) was successfully expressed, purified, and crystallized in a variety of ligand-bound states. Here we report six crystal structures of Mtb CysQ, including a ligand-free structure, a lithium-inhibited state with substrate PAP bound, and a product-bound complex with AMP, phosphate, and three Mg(2+) ions bound. Comparison of these structures together with homologues of the superfamily has provided insight into substrate specificity, metal coordination, and catalytic mechanism.


Subject(s)
Adenosine Diphosphate/metabolism , Bacterial Proteins/chemistry , Mycobacterium tuberculosis/enzymology , N-Glycosyl Hydrolases/chemistry , Adenosine Diphosphate/chemistry , Adenosine Monophosphate/metabolism , Bacterial Proteins/metabolism , Catalysis , Coordination Complexes/chemistry , Coordination Complexes/metabolism , Crystallography, X-Ray , Magnesium/metabolism , Models, Molecular , N-Glycosyl Hydrolases/isolation & purification , N-Glycosyl Hydrolases/metabolism , Phosphates/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Substrate Specificity
11.
Biochim Biophys Acta ; 1837(2): 326-34, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24296033

ABSTRACT

The genome of Saccharomyces cerevisiae contains 35 members of the mitochondrial carrier family, nearly all of which have been functionally characterized. In this study, the identification of the mitochondrial carrier for adenosine 5'-phosphosulfate (APS) is described. The corresponding gene (YPR011c) was overexpressed in bacteria. The purified protein was reconstituted into phospholipid vesicles and its transport properties and kinetic parameters were characterized. It transported APS, 3'-phospho-adenosine 5'-phosphosulfate, sulfate and phosphate almost exclusively by a counter-exchange mechanism. Transport was saturable and inhibited by bongkrekic acid and other inhibitors. To investigate the physiological significance of this carrier in S. cerevisiae, mutants were subjected to thermal shock at 45Ā°C in the presence of sulfate and in the absence of methionine. At 45Ā°C cells lacking YPR011c, engineered cells (in which APS is produced only in mitochondria) and more so the latter cells, in which the exit of mitochondrial APS is prevented by the absence of YPR011cp, were less thermotolerant. Moreover, at the same temperature all these cells contained less methionine and total glutathione than wild-type cells. Our results show that S. cerevisiae mitochondria are equipped with a transporter for APS and that YPR011cp-mediated mitochondrial transport of APS occurs in S. cerevisiae under thermal stress conditions.


Subject(s)
Adenosine Phosphosulfate/metabolism , Genes, Fungal/genetics , Mitochondria/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Adaptation, Physiological , Biological Transport/genetics , Coenzyme A/metabolism , Escherichia coli/metabolism , Genetic Complementation Test , Glutathione/metabolism , Kinetics , Methionine/metabolism , Mutant Proteins/metabolism , Recombinant Proteins/metabolism , Substrate Specificity , Temperature
12.
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
13.
Plant Cell ; 24(10): 4187-204, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23085732

ABSTRACT

3'-Phosphoadenosine 5'-phosphosulfate (PAPS) is the high-energy sulfate donor for sulfation reactions. Plants produce some PAPS in the cytosol, but it is predominantly produced in plastids. Accordingly, PAPS has to be provided by plastids to serve as a substrate for sulfotransferase reactions in the cytosol and the Golgi apparatus. We present several lines of evidence that the recently described Arabidopsis thaliana thylakoid ADP/ATP carrier TAAC transports PAPS across the plastid envelope and thus fulfills an additional function of high physiological relevance. Transport studies using the recombinant protein revealed that it favors PAPS, 3'-phosphoadenosine 5'-phosphate, and ATP as substrates; thus, we named it PAPST1. The protein could be detected both in the plastid envelope membrane and in thylakoids, and it is present in plastids of autotrophic and heterotrophic tissues. TAAC/PAPST1 belongs to the mitochondrial carrier family in contrast with the known animal PAPS transporters, which are members of the nucleotide-sugar transporter family. The expression of the PAPST1 gene is regulated by the same MYB transcription factors also regulating the biosynthesis of sulfated secondary metabolites, glucosinolates. Molecular and physiological analyses of papst1 mutant plants indicate that PAPST1 is involved in several aspects of sulfur metabolism, including the biosynthesis of thiols, glucosinolates, and phytosulfokines.


Subject(s)
Antiporters/physiology , Arabidopsis Proteins/physiology , Arabidopsis/metabolism , Cytosol/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Thylakoids/metabolism , Antiporters/genetics , Antiporters/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Biological Transport , Phosphoadenosine Phosphosulfate/biosynthesis , Plastids/metabolism
14.
Appl Microbiol Biotechnol ; 99(24): 10771-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26293337

ABSTRACT

3'-Phosphoadenosine-5'-phosphosulfate (PAPS) is the obligate cosubstrate and source of the sulfonate group in the chemoenzymatic synthesis of heparin, a clinically used anticoagulant drug. Previously, we have developed a method to synthesize PAPS with Escherichia coli crude extracts, which include three overexpressed enzymes and a fourth unidentified protein. The unknown protein degrades adenosine diphosphate (ADP), the by-product of PAPS synthesis reaction. To further understand and control the process of in vitro enzymatic PAPS synthesis, we decide to identify the fourth protein and develop a defined method to synthesize PAPS using purified enzymes. Here, we show that the purified Nudix hydrolase NudJ degrades ADP at high efficiency and serves as the fourth enzyme in PAPS synthesis. Under the defined condition of PAPS synthesis, all of the 10-mM ADP is hydrolyzed to form adenosine monophosphate (AMP) in a 15-min reaction. ADP is a better substrate for NudJ than adenosine triphosphate (ATP). Most importantly, the purified NudJ does not cleave the product PAPS. The removal of ADP makes the PAPS peak more separable from other components in the chromatographic purification process. This developed enzymatic approach of PAPS production will contribute to the chemoenzymatic synthesis of heparin.


Subject(s)
Adenosine Diphosphate/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Pyrophosphatases/metabolism , Hydrolysis , Nudix Hydrolases
15.
Biochemistry ; 53(3): 591-600, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24401123

ABSTRACT

In bacteria, 3',5'-adenosine bisphosphate (pAp) is generated from 3'-phosphoadenosine 5'-phosphosulfate in the sulfate assimilation pathway, and from coenzyme A by the transfer of the phosphopantetheine group to the acyl-carrier protein. pAp is subsequently hydrolyzed to 5'-AMP and orthophosphate, and this reaction has been shown to be important for superoxide stress tolerance. Herein, we report the discovery of the first instance of an enzyme from the amidohydrolase superfamily that is capable of hydrolyzing pAp. Crystal structures of Cv1693 from Chromobacterium violaceum have been determined to a resolution of 1.9 ƅ with AMP and orthophosphate bound in the active site. The enzyme has a trinuclear metal center in the active site with three Mn(2+) ions. This enzyme (Cv1693) belongs to the Cluster of Orthologous Groups cog0613 from the polymerase and histidinol phosphatase family of enzymes. The values of kcat and kcat/Km for the hydrolysis of pAp are 22 s(-1) and 1.4 Ɨ 10(6) M(-1) s(-1), respectively. The enzyme is promiscuous and is able to hydrolyze other 3',5'-bisphosphonucleotides (pGp, pCp, pUp, and pIp) and 2'-deoxynucleotides with comparable catalytic efficiency. The enzyme is capable of hydrolyzing short oligonucleotides (pdA)5, albeit at rates much lower than that of pAp. Enzymes from two other enzyme families have previously been found to hydrolyze pAp at physiologically significant rates. These enzymes include CysQ from Escherichia coli (cog1218) and YtqI/NrnA from Bacillus subtilis (cog0618). Identification of the functional homologues to the experimentally verified pAp phosphatases from cog0613, cog1218, and cog0618 suggests that there is relatively little overlap of enzymes with this function in sequenced bacterial genomes.


Subject(s)
Adenosine Diphosphate/metabolism , Amidohydrolases/metabolism , Bacterial Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Adenosine Diphosphate/biosynthesis , Amino Acid Sequence , Bacterial Proteins/isolation & purification , Chromobacterium/enzymology , Crystallization , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Phosphoadenosine Phosphosulfate/metabolism , Phosphoric Monoester Hydrolases/isolation & purification , Sequence Alignment , Substrate Specificity
16.
Microbiology (Reading) ; 160(Pt 9): 1832-1844, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24934621

ABSTRACT

The enterobacterium Escherichia coli can utilize a variety of molecules as sulfur sources, including cysteine, sulfate, thiosulfate and organosulfonates. An intermediate of the sulfate assimilation pathway, adenosine 59-phosphosulfate (APS), also acts as a signal molecule regulating the utilization of different sulfur sources. In this work, we show that inactivation of the cysH gene, leading to accumulation of phosphoadenosine 59-phosphosulfate (PAPS), also an intermediate of the sulfate assimilation pathway, results in increased surface adhesion and cell aggregation by activating the expression of the curli-encoding csgBAC operon. In contrast, curli production was unaffected by the inactivation of any other gene belonging to the sulfate assimilation pathway. Overexpression of the cysH gene downregulated csgBAC transcription, further suggesting a link between intracellular PAPS levels and curli gene expression. In addition to curli components, the Flu, OmpX and Slp proteins were also found in increased amounts in the outer membrane compartment of the cysH mutant; deletion of the corresponding genes suggested that these proteins also contribute to surface adhesion and cell surface properties in this strain. Our results indicate that, similar to APS, PAPS also acts as a signal molecule, albeit with a distinct mechanism and role: whilst APS regulates organosulfonate utilization, PAPS would couple availability of sulfur sources to remodulation of the cell surface, as part of a more global effect on cell physiology.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Gene Expression Regulation, Bacterial , Oxidoreductases/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Signal Transduction , Escherichia coli Proteins/genetics , Gene Deletion , Operon , Oxidoreductases/genetics
17.
Semin Cell Dev Biol ; 22(6): 653-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21419855

ABSTRACT

Sulfate (SO(4)(2-)) is an important nutrient for human growth and development, and is obtained from the diet and the intra-cellular metabolism of sulfur-containing amino acids, including methionine and cysteine. During pregnancy, fetal tissues have a limited capacity to produce sulfate, and rely on sulfate obtained from the maternal circulation. Sulfate enters and exits placental and fetal cells via transporters on the plasma membrane, which maintain a sufficient intracellular supply of sulfate and its universal sulfonate donor 3'-phosphoadenosine 5'-phosphosulfate (PAPS) for sulfate conjugation (sulfonation) reactions to function effectively. Sulfotransferases mediate sulfonation of numerous endogenous compounds, including proteins and steroids, which biotransforms their biological activities. In addition, sulfonation of proteoglycans is important for maintaining normal structure and development of tissues, as shown for reduced sulfonation of cartilage proteoglycans that leads to developmental dwarfism disorders and four different osteochondrodysplasias (diastrophic dysplasia, atelosteogenesis type II, achondrogenesis type IB and multiple epiphyseal dysplasia). The removal of sulfate via sulfatases is an important step in proteoglycan degradation, and defects in several sulfatases are linked to perturbed fetal bone development, including mesomelia-synostoses syndrome and chondrodysplasia punctata 1. In recent years, interest in sulfate and its role in developmental biology has expanded following the characterisation of sulfate transporters, sulfotransferases and sulfatases and their involvement in fetal growth. This review will focus on the physiological roles of sulfate in fetal development, with links to human and animal pathophysiologies.


Subject(s)
Cartilage/metabolism , Developmental Biology , Fetal Development/physiology , Infant, Newborn, Diseases/metabolism , Osteochondrodysplasias/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Sulfates/metabolism , Adult , Cartilage/embryology , Cartilage/physiopathology , Child , Cysteine/metabolism , Embryo, Mammalian , Female , Fetus , Humans , Infant, Newborn , Infant, Newborn, Diseases/physiopathology , Membrane Transport Proteins/metabolism , Methionine/metabolism , Osteochondrodysplasias/physiopathology , Pregnancy , Proteoglycans/metabolism , Sulfatases/metabolism , Sulfate Transporters , Sulfotransferases/metabolism
18.
Chem Res Toxicol ; 26(3): 410-21, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23387501

ABSTRACT

Nevirapine (NVP) treatment is associated with serious skin rashes that appear to be immune-mediated. We previously developed a rat model of this skin rash that is immune-mediated and is very similar to the rash in humans. Treatment of rats with the major NVP metabolite, 12-OH-NVP, also caused the rash. Most idiosyncratic drug reactions are caused by reactive metabolites; 12-OH-NVP forms a benzylic sulfate, which was detected in the blood of animals treated with NVP or 12-OH-NVP. This sulfate is presumably formed in the liver; however, the skin also has significant sulfotransferase activity. In this study, we used a serum against NVP to detect covalent binding in the skin of rats. There was a large artifact band in immunoblots of whole skin homogenates that interfered with detection of covalent binding; however, when the skin was separated into dermal and epidermal fractions, covalent binding was clearly present in the epidermis, which is also the location of sulfotransferases. In contrast to rats, treatment of mice with NVP did not result in covalent binding in the skin or skin rash. Although the reaction of 12-OH-NVP sulfate with nucleophiles such as glutathione is slow, incubation of this sulfate with homogenized human and rat skin led to extensive covalent binding. Incubations of 12-OH-NVP with the soluble fraction from a 9,000g centrifugation (S9) of rat or human skin homogenate in the presence of 3'-phosphoadenosine-5'-phosphosulfate (PAPS) produced extensive covalent binding, but no covalent binding was detected with mouse skin S9, which suggests that the reason mice do not develop a rash is that they lack the required sulfotransferase. This is the first study to report covalent binding of NVP to rat and human skin. These data provide strong evidence that covalent binding of NVP in the skin is due to 12-OH-NVP sulfate, which is likely responsible for NVP-induced skin rash. Sulfation may represent a bioactivation pathway for other drugs that cause a skin rash.


Subject(s)
Exanthema/chemically induced , Nevirapine/adverse effects , Nevirapine/metabolism , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/metabolism , Skin/metabolism , Animals , Exanthema/metabolism , Exanthema/pathology , Female , HIV Infections/drug therapy , Humans , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NADP/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Protein Binding , Proteins/metabolism , Rats , Skin/pathology
19.
ACS Synth Biol ; 12(5): 1487-1496, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37042633

ABSTRACT

3'-Phosphoadenosine-5'-phosphosulfate (PAPS) is the bioactive form of sulfate and is involved in all biological sulfation reactions. The enzymatic transformation method for PAPS is promising, but the low efficiency and high cost of enzyme purification and storage restrict its practical applications. Here, we reported PAPS biosynthesis with a protein crystalline inclusion (PCI)-based enzyme immobilization system. First, the in vivo crystalline inclusion protein CipA was identified as an efficient auto-assembly tag for immobilizing the bifunctional PAPS synthase (ASAK). After characterizing the pyrophosphokinase activity of a polyphosphate exonuclease PaPPX from Pseudomonas aeruginosa, and optimizing the linker fragment, auto-assembled enzymes ASAK-PT-CipA and PaPPX-PT-CipA were constructed. Then, the auto-assembled enzymes ASAK-PT-CipA and PaPPX-PT-CipA with high stability were co-expressed and immobilized for constructing a transformation system. The highest transformation rate of PAPS from ATP and sulfate reached 90%, and the immobilized enzyme can be reused 10 times. The present work provided a convenient, efficient, and easy to be enlarged auto-immobilization system for PAPS biosynthesis from ATP and sulfate. The immobilization system also represented a new approach to reduce the production cost of PAPS by facilitating the purification, storage, and reuse of related enzymes, and it would boost the studies on biotechnological production of glycosaminoglycans and sulfur-containing natural compounds.


Subject(s)
Enzymes, Immobilized , Sulfate Adenylyltransferase , Sulfate Adenylyltransferase/genetics , Sulfate Adenylyltransferase/chemistry , Sulfate Adenylyltransferase/metabolism , Sulfates/metabolism , Phosphoadenosine Phosphosulfate/metabolism , Adenosine Triphosphate/metabolism
20.
Methods Mol Biol ; 2557: 709-720, 2023.
Article in English | MEDLINE | ID: mdl-36512246

ABSTRACT

Subcellular fractionation is an introductory step in a variety of experimental approaches designed to study intracellular components, like membranes and organelle systems. Subcellular fractions enriched in membranes of the Golgi apparatus of mammalian cells have been isolated to address localization and activity of proteins, including enzymes, to study intracellular membrane transport mechanisms, and to reconstitute in vitro cellular processes associated with the Golgi apparatus. Here, I describe methods to purify Golgi membranes by subcellular fractionation, to assay nucleotide sulfate (PAPS) uptake into Golgi vesicles, and to measure sulfate incorporation into in vitro synthesized glycosaminoglycans.


Subject(s)
Phosphoadenosine Phosphosulfate , Proteoglycans , Animals , Phosphoadenosine Phosphosulfate/metabolism , Proteoglycans/metabolism , Golgi Apparatus/metabolism , Glycosaminoglycans/metabolism , Sulfates/metabolism , Mammals/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL