Biology and function of the reversible sulfation pathway catalysed by human sulfotransferases and sulfatases

https://doi.org/10.1016/S0009-2797(97)00117-8Get rights and content

Abstract

Sulfation and sulfate conjugate hydrolysis play an important role in metabolism, and are catalysed by members of the sulfotransferase and sulfatase enzyme super-families. In general, sulfation is a deactivating, detoxication pathway, but for some chemicals the sulfate conjugates are much more reactive than the parent compound. The range of compounds which are sulfated is enormous, yet we still understand relatively little of the function of this pathway. This review summarises current knowledge of the sulfation system and the enzymes involved, and illustrates how heterologous expression of sulfotransferases (SULTs) and sulfatases is aiding our appreciation of the properties of these important proteins. The role of sulfation in the bioactivation of procarcinogens and promutagens is discussed, and new data on the inhibition of the sulfotransferase(s) involved by common dietary components such as tea and coffee are presented. The genetic and environmental factors which are known to influence the activity and expression of human SULTs and sulfatases are also reviewed.

Introduction

Sulfation1 has evolved as a key step in xenobiotic metabolism, but also plays a critical role in steroid biosynthesis and in modulating the biological activity and facilitating the inactivation and elimination of potent endogenous chemicals including thyroid hormones, steroids and catechols 1, 2, 3. The sulfation pathway can be thought of as reversible, comprising two enzyme systems: the sulfotransferases (SULTs) which catalyse the sulfation reaction and the sulfatases which catalyse the hydrolysis of sulfate esters formed by the action of SULTs. These two enzyme systems are both the products of multigene families, with the SULTs located in the cytoplasm and the arylsulfatases (ARS) present in either the lysosomes or the endoplasmic reticulum. Although much is now known of the molecular biology of these enzyme systems, less is understood about their detailed biological function, and in particular about how the interplay between various members of the SULT and ARS enzyme families regulates the availability and biological activity of xenobiotics and endogenous chemicals which constitute their substrate profiles. Of particular interest and relevance is how genetic, hormonal and environmental factors control the relative isoenzyme complements in different tissues and at different stages of development and ageing.

This review will focus on the various essential components of the sulfation systems and on some of the studies underway to try and understand the function of these fascinating and important enzymes.

Section snippets

Components of the sulfation system

When considering the biological function of sulfation, it is important to recognise the various components which constitute the whole sulfation system, not just the enzymes themselves, since the individual components will contribute collectively to the overall capacity and ability for sulfation in any given in vitro or in vivo system. Key features of the system are the supply of substrate, the supply of the activated sulfate donor molecule (or co-substrate) PAPS, and the balance between

Supply of substrate

The supply of substrate for the enzymes of sulfation is obviously of central importance to the overall sulfation system. Absorption of xenobiotics through the skin, lungs or gastrointestinal tract, transport in the circulation, import or export to and from target cells and intracellular movement of small molecules will collectively determine the availability of compounds for the reversible sulfation pathway. Sulfate conjugates are, in general, polar and water soluble molecules, which should not

Supply of PAPS

3′-phosphoadenosine 5′-phosphosulfate or 3′-phosphoadenylylsulfate (PAPS) is the sulfate donor molecule for eukaryotic SULTS. PAPS is formed from ATP and inorganic sulfate by two enzyme activities, ATP sulfurylase and APS kinase. In the first step Eq. (1), adenosine 5′-phosphosulfate is formed by the action of ATP sulfurylase and as a result of the kinetically unfavourable nature of this reaction in the forward (physiological) direction, APS kinase Eq. (2)drives the activation pathway for

Sulfotransferase enzymes

The human SULT enzyme family as currently known comprises of five well characterised members, each the product of a distinct gene, with widely differing substrate specificities and amino acid sequences [3]. Based on amino acid sequence identity and substrate preferences, the various human SULTs fully characterised to date can be subdivided into two families, phenol SULTs (P-PST, SULT1A2, M-PST and EST) and hydroxysteroid SULT (HST). The existence of further human SULTs, related to the rat SULTs

Arylsulfatase enzymes

The arylsulfatase family was classically thought to comprise of three members: the lysosomal ARSA and ARSB which hydrolyse sulfates of glycolipids and complex carbohydrates such as dermatan and chondroitin, respectively, and the steroid sulfatase, or arylsulfatase C (ARSC), which is present in the endoplasmic reticulum and which hydrolyses sulfate esters of a wide range of steroids and cholesterol (Table 3). Sulfate conjugates of xenobiotics are also hydrolysed by ARS. These ARS enzymes and

Heterologous expression systems for human SULT and ARS enzymes

The ability to study the kinetic and catalytic properties of individual SULT and ARS enzymes in isolation is extremely valuable for understanding their substrate specificities, and also the physical and chemical properties of enzyme and substrate which determine substrate preference. Heterologous expression allows the synthesis of recombinant enzyme protein from a cDNA clone in, usually, bacterial, yeast, insect or mammalian cells. The properties of the enzyme (e.g. membrane bound, cytosolic,

Biological function of sulfation

Sulfation of small molecules appears ubiquitous to mammalian species, however, we are only beginning to unravel the complexities of its functions. Based on current knowledge, the functions of sulfation may be sub-divided into the broad areas of chemical defence, hormone biosynthesis and bioactivation.

Sulfation and chemical defence

Sulfation is considered an important component of what is called phase II of xenobiotic metabolism, i.e. the conjugation and true detoxication step and is, therefore, a central feature of the body’s chemical defence armoury. The classical view of sulfation is that it protects against the toxic or potentially toxic effects of numerous xenobiotics and their metabolites, as sulfate conjugates are in general more polar, more water soluble and, therefore, more readily excreted in urine or bile, in

Sulfation and steroid biosynthesis

In humans dehydroepiandrosterone sulfate (DHEAS) is the major circulating steroid 60, 61and estrone sulfate is the major estrogen in the circulation [62]. These chemicals are known to act as precursors for the formation of estrogens and androgens. For example, during the second half of pregnancy, the human fetal adrenal gland secretes extremely large quantities of DHEAS which is utilised (following hydroxylation by cytochrome P450 and hydrolysis by steroid sulfatase, ARSC) as the major

Sulfation as a bioactivation reaction

It is increasingly recognised that whilst sulfation is normally a detoxication reaction, with the sulfate conjugate exhibiting reduced biological and/or pharmacological activity compared with the parent compound, there are a number of important examples where sulfate conjugates are more biologically active than the corresponding free compounds.

Drugs such as minoxidil 70, 71and cicletanine [72]are bioactivated upon sulfation, with the sulfate conjugate identified as the pharmacologically active

Dietary compound interaction with sulfation

Most SULTs are expressed (to varying degrees) in the human gastrointestinal tract 52, 86, 87, 88, 89. In light of this fact and of the important role sulfation plays in the bioactivation of dietary procarcinogens, it is reasonable to assume that there is potential for the production of highly reactive, unstable, mutagenic sulfuric acid esters within the gastrointestinal tract. This may be an important mechanism in, for example, colon cancer where dietary aromatic amines known to be activated

Genetic and environmental factors affecting sulfation

The important biological functions of sulfation outlined above may be adversely affected by factors influencing the expression and/or activity of components of the sulfation system. The effect of defective PAPS synthesis is illustrated by the brachymorphic mouse 9, 81, 82. Additional factors known to affect sulfation are described below.

Sulfatases

There are a number of genetic diseases associated with heritable mutations in the genes coding for ARSA, ARSB, ARSC 21, 22, 23and ARSE [24], as well as the multiple sulfatase deficiency which affects all sulfatases 22, 26(Table 3). Of these, X-linked ichthyosis is probably the most pertinent to small molecule sulfation.

X-linked ichthyosis, which results from mutations (usually complete gene deletions) in the STS gene coding for steroid sulfatase or ARSC, is one of the more common genetic

Sulfotransferases

To date, no genetic disease associated with a defect in any cytosolic sulfotransferase is known. However, there is extreme variation in the expression of various SULTs within the human population and there is good evidence for the existence for a genetic polymorphism affecting the P-PST isoform 3, 117, 118. HST expression in human liver also appears to follow a bimodal distribution, with about 25% of individuals belonging to a high activity group [119]. The explosion in the cloning of the cDNAs

Conclusions

Over the last 10 years or so, much progress has been made in our understanding of the sulfation system. The application of recombinant DNA technology has rapidly advanced our ability to identify, catalogue and characterise the members of the SULT and ARS gene families although it is clear that additional members remain to be identified. Heterologous expression in prokaryotic and eukaryotic systems has facilitated our study of their substrate specificities and provides the raw material for

Acknowledgements

Recent work in our laboratory has been supported by Action Research, the Biotechnology and Biological Sciences Research Council, the Commission of the European Communities (BMH1-CT92-0097, EV5V-CT94-0410), Glaxo Wellcome, the Scottish Hospitals Endowment Research Trust and the Wellcome Trust. cDNAs encoding ARSA, ARSB and ARSC were generously provided by Professor Kurt von Figura.

References (129)

  • X Zhu et al.

    cDNA cloning and expression of a new form of human aryl sulfotransferase

    Int. J. Biochem. Cell. Biol.

    (1996)
  • I.A Aksoy et al.

    Human liver estogen sulfotransferase: identification by cDNA cloning and expression

    Biochem. Biophys. Res. Commun.

    (1994)
  • T.C Wood et al.

    Human liver thermolabile phenol sulfotransferase, cDNA cloning, expression and characterization

    Biochem. Biophys. Res. Commun.

    (1994)
  • A.L Jones et al.

    Human platelet phenol-SULTs, cDNA cloning, stable expression in V79 cells and identification of a novel allelic variant of the phenol-sulfating form

    Biochem. Biophys. Res. Commun.

    (1995)
  • K.J Forbes et al.

    Human fetal adrenal hydroxysteroid sulphotransferase: cDNA cloning stable expression in V79 cells and functional characterisation of the expressed enzyme

    Mol. Cell. Endocrinol.

    (1995)
  • R.P Remmel et al.

    Validation and use of cloned, expressed human drug-metabolizing enzymes in heterologous cells for analysis of drug metabolism and drug-drug interactions

    Biochem. Pharmacol.

    (1993)
  • H Glatt et al.

    Activation of promutagens by endogenous and heterologous SULTs expressed in continuous cell-cultures

    Toxicol. Lett.

    (1994)
  • H Glatt et al.

    Activation of benzylic alcohols to mutagens by rat and human SULTs expressed in E. coli

    Eur. J. Pharmacol.

    (1995)
  • H Glatt et al.

    S. typhimurium strains and mammalian cells genetically engineered for expression of sulfotransferases

    Toxicol. Lett.

    (1995)
  • C Stein et al.

    Cloning and expression of human steroid sulfatase. Membrane topology glycosylation and subcellular distribution in BHK-21 cells

    J. Biol. Chem.

    (1989)
  • C Peters et al.

    Phylogenetic conservation of ARS-cDNA cloning and expression of human arylsulfatase B

    J. Biol. Chem.

    (1990)
  • C Stein et al.

    Cloning and expression of human arylsulfatase A

    J. Biol. Chem.

    (1989)
  • D Ratge et al.

    Plasma free and conjugated catecholamines in clinical disorders

    Life Sci.

    (1986)
  • B.J Buirchell et al.

    Metabolism of estradiol-17β in human endometrium during the menstrual cycle

    J. Steroid Biochem.

    (1975)
  • A.L Jones et al.

    Estrogen and phenol sulfotransferase activities in human fetal lung

    Early Hum. Dev.

    (1992)
  • M.N Woods et al.

    Low-fat, high-fiber diet and serum estrone sulfate in premenopausal women

    Am. J. Hum. Nutr.

    (1989)
  • K.W Selcer et al.

    Inhibition of placental estrone sulfatase activity and MCF-7 breast cancer cell proliferation by estrone-3-amino derivatives

    J. Steroid Biochem. Mol. Biol.

    (1996)
  • J.I Rearick et al.

    Increase in cholesterol sulfotransferase activity during in vitro squamous differentiation of rabbit tracheal epithelial cells and its inhibition by retinoic acid

    J. Biol. Chem.

    (1987)
  • A.M Jetten et al.

    Increased cholesterol sulfate and cholesterol sulfotransferase activity in relation to the multi-step process of differentiation in human epidermal keratinocytes

    J. Invest. Dermatol.

    (1989)
  • K.D Meisheri et al.

    Enzymatic and non-enzymatic sulfation mechanisms in the biological actions of minoxidil

    Biochem. Pharmacol.

    (1993)
  • R.P Garay et al.

    Evidence for (+)-cicletanine sulfate as an active natriuretic metabolite of cicletanine in the rat

    Eur. J. Pharmacol.

    (1995)
  • P Robel et al.

    Neurosteroids: biosynthesis and function

    Trends Endocrinol. Metab.

    (1994)
  • J.J Lambert et al.

    Neurosteroids and GABAA receptor function

    Trends Pharmacol. Sci.

    (1995)
  • P Li et al.

    Memory enhancement mediated by the steroids sulfatase inhibitor (p-O-sulfamoyl)-N-tetradecanoyl tyramine

    Life Sci.

    (1996)
  • P Robel et al.

    Biosynthesis and assay of neurosteroids in rats and mice: functional correlates

    J. Steroid Biochem. Mol. Biol.

    (1995)
  • J.A Miller

    Sulfonation in chemical carcinogenesis, history and present status

    Chem.–Biol. Interact.

    (1994)
  • K Sugahara et al.

    Defect in PAPS synthesis in brachymorphic mice. I. Characterization of the defect

    Arch. Biochem. Biophys.

    (1982)
  • K Sugahara et al.

    Defect in PAPS synthesis in brachymorphic mice. II. Tissue distribution of the defect

    Arch. Biochem. Biophys.

    (1982)
  • G Rein et al.

    Mutiple forms of phenolsulphotransferase in human tissues. Selective inhibition by dichloronitrophenol

    Biochem. Pharmacol.

    (1982)
  • A Baranczyk-Kuzma et al.

    Human ileum phenol sulfotransferase

    Biochem. Pharmacol.

    (1989)
  • Z Apostolides et al.

    Inhibition of PhlP mutagenicity by catechins and by theaflavins and gallate esters

    Mutat. Res.

    (1997)
  • B Stavric

    Quercetin in our diet, from potent mutagen to probable anticarcinogen

    Clin. Biochem.

    (1994)
  • J.H Weisburger et al.

    Tea polyphenols as inhibitors of mutagenicity of major classes of carcinogens

    Mutat. Res.

    (1996)
  • D.H Barch et al.

    Dietary ellagic acid inhibits the enzymatic activity of CYP1A1 without altering hepatic concentrations of CYP1A1 or CYP1A1 messenger RNA

    Biochem. Biophys. Res. Commun.

    (1994)
  • P Talalay et al.

    Chemoprotection against cancer by phase-2 enzyme induction

    Toxicol. Lett.

    (1995)
  • C Gibb et al.

    In vitro inhibition of phenolsulphotransferae by food and drink constituents

    Biochem. Pharmacol.

    (1987)
  • R Weinshilboum et al.

    Sulfotransferase enzymes

  • M.W.H Coughtrie

    Sulphation catalysed by the human cytosolic sulphotransferases-chemical defence or molecular terrorism?

    Hum. Exp. Toxicol.

    (1996)
  • G Jedlitschky et al.

    Transport of glutathione, glucuronate and sulfate conjugates by the MRP gene-encoded conjugate export pump

    Cancer Res.

    (1996)
  • A.M Hassen et al.

    Uptake of sulfate conjugates by isolated rat hepatocytes

    Drug Metab. Dispos.

    (1996)
  • Cited by (197)

    • Genetics and pathophysiology of mammalian sulfate biology

      2017, Journal of Genetics and Genomics
      Citation Excerpt :

      Cartilage defects and lethal phenotypes have been linked to Slc35b2 in Zebrafish and Drosophila (Kamiyama et al., 2003; Clément et al., 2008), whereas up-regulation of SLC35B2 mRNA expression is associated with poor prognosis of invasive ductal breast carcinoma in humans (Chim-ong et al., 2014). The landmark paper reporting sulfate activation to PAPS (Lipmann, 1958) and the following identification of sulfotransferases (Lipmann, 1958; Gamage et al., 2006), as well as the characterisation of animal models of perturbed sulfate homeostasis (Tables 5–7), have led to our current understanding of sulfate biology genetics, and the physiological importance of sulfate in modulating the biological activity of neurotransmitters (Coughtrie et al., 1998; Lee et al., 2007), steroids (Dawson, 2012), thyroid hormone (Richard et al., 2001; Wu et al., 2005), proteoglycans and xenobiotics (Habuchi et al., 2004; Klüppel, 2010). Sulfotransferases, can be grouped into two classes: (i) SULTs which sulfonate neurotransmitters, bile acids, xenobiotics and steroids; (ii) Golgi-located STs that have proteoglycan and lipid substrates (Gamage et al., 2006).

    View all citing articles on Scopus
    View full text