2,129
Views
10
CrossRef citations to date
0
Altmetric
Research Article

A microtitre plate assay for measuring glycosidase activity

, , , , , , , & show all
Pages 131-135 | Received 15 Jan 2007, Accepted 03 Mar 2007, Published online: 04 Oct 2008

Abstract

Glycosidases perform a wide range of functions in physiology and pathology, and are potential targets for the treatment of diseases such as influenza, cancer, AIDS and diabetes. This paper reports a convenient discontinuous colourimetric assay for the measurement of glycosidase activity. The assay utilises 4-nitrophenyl- substrates and quantities of product are determined by measuring absorbance at 405 nm. This assay is performed in a 96 well microtitre plate and has been used to characterise the properties of seven different glycosidases from bacteria, yeast and higher eukaryotes and their kinetic parameters determined. Assays in the presence of known inhibitors showed that inhibition modes can be determined, and IC50 and Ki values calculated. This assay appears to be of widely applicable and of general utility for the measurement of glycosidase activity and the evaluation of inhibitors.

Introduction

Sugar conjugates are ubiquitous and found within virtually all prokaryotes and eukaryotes. Sugars can be conjugated to other sugars, amino acids (especially in proteins), steroid hormones and other molecules. Examples of these conjugates include glycogen, starch and other polysaccharides, glycoproteins, natural glycosides such as salicilin and digitoxin and the glucuronate conjugates of hormones, drugs and other xenobiotics. The ubiquitous nature of these glycoside conjugates means that all organisms require enzymes for their synthesis and degradation.

Glycosidase enzymes catalyse the hydrolysis of sugar conjugates resulting in breaking of the glycosidic bond. Glycosides are ubiquitous and many glycosidases have the potential to be drug targets. Important examples of glycosidases include the influenza virus neuramidase [Citation1], and the Williopsis saturnus killer toxin which has potent anti-microbial activity [Citation2]. Glycosidases are important potential drug targets and in some cases therapies have been successfully developed. The influenza anti-viral agents Relenza™ and Tamiflu™ are inhibitors of the viral neuramidase, and have been approved for use in most of the world [Citation1]. Increased activity of β-glucuronidase has been implicated in increased hormone levels in breast and some other cancers Citation3-5, and this is thought to be due to decreased excretion of the hormone by glucuronidation. Glucuronidase inhibitors, such as glucaro-1,4-lactone, have been shown to control carcinogenesis in animal models [Citation6]. α-Glucosidases may be of value in treating diabetes and AIDS, whilst α-mannosidase inhibitors inhibit cancer metastasis [Citation7].

Given the importance of glycosidase enzymes, several papers have reported continuous or discontinuous colourimetric assays using chromogenic substrates Citation8-17. Surprisingly, there appears to be no general evaluation and validation of these assay methods, and many of the reported methods are unsuitable for kinetic analyses of the enzymes or characterisation of inhibitors. This paper addresses this omission by studying seven different galactosidases, glucosidases and glucuronidases from bacteria, yeast, and higher eukaryotic sources.

Materials and methods

Materials

All reagents were supplied by the Sigma-Aldrich Chemical Co. or Fisher Ltd unless otherwise specified and were used without further purification. MOPS1 buffer was supplied by BDH. 4-Nitrophenyl- substrates were supplied by Fluka. All reagents were prepared in 18 Ω Milli-Q water (Millipore). Buffer solutions were pH adjusted with NaOH or HCl using a Stuart Scientific pH meter calibrated with pH 7.0 and 4.0 or 10.0 standards (BDH).

Activity assays

The following commercial enzymes (Sigma) were used in this study: β-D-Galactosidase from Aspergillus oryzae (G5160; E.C. 3.2.1.23); β-D-Galactosidase from E. coli (G2513; E.C. 3.2.1.23); α-D-glucosidase from S. cerevisiae (63412; E.C. 3.2.1.20); β-D-glucosidase from Amygdalae dulces (G4511; E.C. 3.2.1.21); β-D-Glucuronidase from bovine liver (G0251; E.C. 3.2.1.31); β-D-glucuronidase from E. coli (G7396; E.C. 3.2.1.31); and β-D-glucuronidase from Patella vulgate (keyhole limpet) (G-8132; E.C. 3.2.1.23). One unit of activity is the amount of enzyme producing 1 μmol of product per min except for the glucuronidase enzymes when one unit is the amount of enzyme producing 1 μg phenolphthalein per hour at 37°C, under the conditions defined by the Sigma-Aldrich Chemical Co. The purity of enzymes was analysed using SDS-PAGE [Citation18] using a Mini-Protean III system (Bio-Rad) with a 3% stacking gel and 10% separating gel. Concentrated enzyme stock solutions (100 U/mL for galactosidases and glucosidases, 100 000 U/mL for glucuronidases) were made up in 20 mM buffer at their optimum pH () and stored in aliquots at − 80°C.

Table I.  Assay conditions and kinetic parameters investigated for enzymes used in this study. Product was quantified by reference to a standard curve (see experimental).

Enzyme reaction mixtures were usually conducted in triplicate and consisted of buffer (50 mM final concentration, at the optimum pH for each enzyme), substrate (1 mM final concentration), inhibitor (if required) and water (to a final volume of 1 mL) and were pre-warmed at 37°C for 5 min. Assays were initiated using ∼50 μL of enzyme in 20 mM buffer (0.2–1.5 U/mL for galactosidases, glucosidases and β-glucuronidase from E. coli; ∼500–1000 U/mL for other glucuronidases) at 10 s intervals and 100 μL aliquots were withdrawn every 2–3 min at the same 10 s intervals for 10–15 min, to ensure that each sample was incubated for exactly the required time. The withdrawn aliquot was quenched in a 96 well microtitre plate containing 100 μL of 100 mM NaOH (assays for β-glucuronidase from P. vulgate required 250 mM NaOH due to the very acidic nature of the buffer) and the absorbance at 405 nm was measured using a VERSAmax tunable microtitre plate reader (Molecular Devices). Negative assay controls contained no substrate, no enzyme, or enzyme boiled at 95°C for 10 min before assaying. Control experiments showed that colour development was stable for at least 30 min after quenching with NaOH. A standard curve was used to quantify product and was made from 4-nitrophenol solution adjusted to pH 12 with NaOH (nmol product = 15.215 A405+0.8028; R2 = 0.9769). The concentration of the stock solution was determined spectrophotometrically assuming ε401nm = 18.1 mM− 1 cm− 1 [Citation19]. Assays for β-glucuronidase using phenolphthalein β-glucuronide as substrate were conducted in an analogous manner except that microtitre plates were analysed at 552 nm. A standard curve of phenolphthalein was constructed assuming ε552nm = 26.6 mM− 1cm− 1 [Citation20]. Protein concentrations were calculated from the U/mg data provided by the Sigma-Aldrich Chemical Co.

Kinetic assays

Assays to determine kinetic parameters were conducted as for general assays except that around 8 different substrate concentrations between 0.1–10 mM final concentration were used. Most substrates are only soluble to ∼40 mM in water, and hence larger volumes of stock solution were added to the assay with adjustment of the volume of water. Kinetic data was analysed by the method of Lineweaver and Burk [Citation21] to ensure adherence to the Michaelis-Menten equation and kinetic parameters estimated using the Direct Linear Plot [Citation22,Citation23]. Assays containing inhibitors were performed either at fixed inhibitor concentration with variable substrate concentrations (as above) or at fixed substrate concentration (at a concentration slightly above the determined Km value) with variable inhibitor concentrations.

Results

Assays for glycosidase activity were initially conducted with the cognate substrate at a fixed concentration of 1 mM (Scheme ). Reactions were carried out over a 10–15 minute time course with variable amounts of enzyme to determine the required quantity. In most cases this was ∼50 μL of enzyme at 0.2–1.5 U/mL (galactosidases, glucosidases and E. coli β-glucuronidase) or 50 μL at 500–1000 U/mL (other glucuronidases) in a 1 mL assay, and all enzymes showed a linear relationship between reaction rate and amount of enzyme added. Control experiments using no enzyme or boiled enzyme showed that the substrates were stable under a wide variety of pH and buffer conditions and under the alkali quenching conditions. Thus, the coloured 4-nitrophenolate product in the assay was exclusively due to enzymatic activity. The product was quantified using a standard curve, and hence rates in units of μmol/min/mg protein can be calculated.

Scheme 1 (A). Reactions catalysed by glycosidases as exemplified by β-glucosidase. Under the alkali quenching conditions 4-nitrophenol (pKa = 7.2) ionises to 4-nitrophenolate to give a yellow colour (ε401nm = 18.5 mM− 1 cm− 1 [Citation11]); (B). Structures of substrates used in this study: 1, 4-nitrophenyl-β-D-glucopyranoside; 2, 4-nitrophenyl-α-D-glucopyranoside; 3, 4-nitrophenyl-β-D-galactopyranoside; 4, 4-nitrophenyl-β-D-glucuronide; 5, phenolphthalein-β-D-glucuronide.

Scheme 1 (A). Reactions catalysed by glycosidases as exemplified by β-glucosidase. Under the alkali quenching conditions 4-nitrophenol (pKa = 7.2) ionises to 4-nitrophenolate to give a yellow colour (ε401nm = 18.5 mM− 1 cm− 1 [Citation11]); (B). Structures of substrates used in this study: 1, 4-nitrophenyl-β-D-glucopyranoside; 2, 4-nitrophenyl-α-D-glucopyranoside; 3, 4-nitrophenyl-β-D-galactopyranoside; 4, 4-nitrophenyl-β-D-glucuronide; 5, phenolphthalein-β-D-glucuronide.

The substrate selectivity of each glycosidase was tested using all of the different substrates (Scheme ) at 1 mM final concentration under the optimised assay conditions. In most cases each enzyme had almost complete selectivity for its preferred substrate. Colour development in assays with other substrates was not significantly higher than for negative controls in most cases, although weak activity was observed for a few enzyme/substrate combinations. Both the 4-nitrophenyl- and phenolphthalein- derivatives of glucuronic acid were substrates for the glucuronidases tested. The phenolphthalein derivative is a previously characterised substrate for these enzymes [Citation20], and in some cases the rate of its conversion was approximately 10-fold higher for it compared to the 4-nitrophenyl- substrate.

Kinetic parameters for each of the enzymes were then determined. For the majority of enzymes Km values were between 0.2 to 2 mM (, 16), but β-glucuronidase from P. vulgate had a lower Km value of 0.052 mM (, 7). Values for Vmax varied more widely (∼0.2–280 μmol/min/mg protein), and this is a reflection of the catalytic efficiency for conversion of the appropriate substrate. Proper Michaelis-Menten kinetics was observed for all the enzymes examined in this study over the range of substrate concentrations utilised. However, previous experiments have shown that some enzymes exhibit non-competitive substrate inhibition in these assays (R. I. Davis and M. D. Lloyd, unpublished results), i.e. rates at higher substrate concentrations are lower than expected.

Experiments using known inhibitors of enzymes indicated that the expected reduction in rate was observed in their presence. Using this assay it was possible to determine IC50 values, e.g. the known suicide inhibitor conduritol-β-epoxide in our study had an IC50 value of 0.57 mM for A. dulces β-glucosidase. The reported Ki value for conduritol-β-epoxide and human β-glucosidase is 166 ± 57 μM [Citation24]. It was also possible to determine inhibition mode and Ki values, e.g. in our study glucuronic acid and gluconolactone were both competitive inhibitors of β-glucuronidase from E. coli, with Ki values of 0.60 mM and 0.49 mM, respectively.

Discussion

Nitrophenyl-glycoside conjugates of sugars were chosen as substrates as they are commercially available, for their stability and the stability of the coloured nitrophenol product under the assay conditions. Our assays used 4-nitrophenyl- conjugates as the extinction coefficient of the 4-nitrophenolate product (18.1 mM− 1 cm− 1 [Citation19]) gives the greatest sensitivity. The assay methods are also applicable to 2-nitrophenol derivatives, but their use will result in lower sensitivity due to the lower extinction coefficient of 2-nitrophenolate (3.5 mM− 1 cm− 1 [Citation25]). The 4-nitrophenyl-glycoside conjugates were substrates for all of the enzymes tested in our study, even those enzymes (β-glucuronidases) traditionally assayed using alternative substrates (phenolphthalein-glucuronide). Phenolphthalein-glucuronide is considerably more expensive than the 4-nitrophenol derivative and colour development in our phenolphthalein-based assays was observed to be unreproducible under our assay conditions, possibly due to tautomerisation of the phenolphthalein product [Citation26]. Thus, 4-nitrophenyl-glucuronide is the first choice as substrate if it is converted by the enzyme under study.

Many of the previously reported assays [Citation8,Citation11,Citation15,Citation16,Citation25] have utilised neutral pH values (∼7.0), and did not adjust the pH before the quantifying product. Since the pKa of 4-nitrophenol/4-nitrophenolate is reported to be 7.14 [Citation19] and the absorbance of 4-nitrophenol is weak (0.2 mM− 1 cm− 1 [Citation19]), this will result in serious under-estimation of the amount of product, assuming no change in the product pKa under the assay conditions. Moreover, this error is likely to be highly variable at neutral pH values, since small changes in pH will lead to relatively large changes in the proportion of the 4-nitrophenolate anion. At acidic pH values the assay would be extremely insensitive because the measured product is 4-nitrophenol not 4-nitrophenolate. To overcome these problems we used a discontinuous assay and quenched under alkali conditions (pH > 12). Our procedure both stops the reaction and ensures full colour development allowing proper product quantification. The procedure worked even for enzymes assayed under very acidic conditions (e.g. β-glucuronidase from P. vulgate, pH 3.8), which could not be assayed using a continuous method. A similar procedure to ours using a 96 well plate [Citation17] has been reported, but this assay was not utilised for kinetic analyses of enzymes.

Unlike previous studies [Citation11], our study shows that this type of assay is applicable to enzymes utilising different glycoside moieties and to enzymes from a variety of prokaryotic and eukaryotic sources. Kinetic parameters could be obtained for all of the enzymes (), but it was necessary to rigorously characterise each enzyme/substrate pair in order to identify appropriate assay conditions. When screening for competitive and mixed inhibitors at fixed substrate concentrations (i.e. to determine IC50 values) a similar or slightly higher substrate concentration to their Km value is appropriate. This is because the characteristic of competitive inhibition is that the Km value apparently increases with consequent decreases in rate and high substrate concentrations will negate this effect. Moreover, non-competitive substrate inhibition is more likely to be observed at high substrate concentrations, thereby increasing the complexity of the analysis. Using known inhibitors of our enzymes we demonstrated that IC50 values, inhibition mode and Ki values can be determined using our assay. Thus, our assays seem to be widely applicable to the measurement of glycosidase enzyme activity, screening for inhibitors and the determination of kinetic and inhibition constants. The current assay system could be adapted to a 96 well plate format relatively easily for the medium throughput determination of IC50 values, although the concentrations of enzyme and substrate will need to be individually optimised.

Abbreviations
MOPS:=

morpholino-N-sulfonic acid

SDS-PAGE:=

sodium dodecyl sulfate polyacrylamide gel electrophoresis

Acknowledgements

We thank Dr P. J. Wood and Dr K. D. E. Boardman (University of Bath, U.K.) for assistence and Dr A. Watts (University of Bath, U.K.) for critically reading this manuscript. This work was funded by a University of Bath undergraduate project grant to MDL.

References

  • Garman E, Laver G. Controlling influenza by inhibiting the virus's neuraminidase. Current Drug Targets 2004; 5: 119–136
  • Guyard C, Dehecq E, Tissier JP, Polonelli L, Dei-Cas E, Cailliez JC, Menozzi FD. Involvement of β-glucans in the wide-spectrum antimicrobial activity of Williopsis saturnus var. mrakii MUCL 41968 killer toxin. Mol Med 2002; 8: 686–694
  • Yuan L, Wagatsuma C, Sun BX, Kim JH, Surh YJ. The role of β-glucuronidase in induction of apoptosis by genistein combined polysaccharide (GCP) in xenogeneic mice bearing human mammary cancer cells in Apoptosis: From Signaling Pathways to Therapeutic Tools. M Diederich, 2003; 347–349
  • Kim DH, Jin YH. Intestinal bacterial β-glucuronidase activity of patients with colon cancer. Arch Pharm Res 2001; 24: 564–567
  • Paigen K, Peterson J, Paigen B. Role of urinary β-glucuronidase in human bladder cancer. Cancer Res 1984; 44: 3620–3623
  • Walaszek Z, Szemraj J, Narog M, Adams AK, Kilgore J, Sherman U, Hanausek M. Metabolism, uptake, and excretion of a D-glucaric acid salt and its potential use in cancer prevention. Cancer Detect Prev 1997; 21: 178–190
  • Jacob GS. Glycosylation inhibitors in biology and medicine. Curr Opin Struct Biol 1995; 5: 605–611
  • Aich S, Delbaere LTJ, Chen R. Continuous spectrophotometric assay for β-glucuronidase. BioTechniques 2001; 30: 846
  • Xu GB, Zhu LH, Hong JM, Cao Y, Xia TA. Rapid colorimetric assay of urinary β-galactosidase and N-acetyl-β-D-glucosaminidase with Cobas Mire auto-analyser. J Clin Lab Anal 1999; 13: 95–98
  • Huidobro JF, Santana FJ, Sanchez MP, Sancho MT, Muniategui S, Simallozano J. J Diastase, invertase and β-glucosidase activities in fresh honey from north-west Spain. Apicultural Res 1995; 34: 39–44
  • Ferreira C, Torres BB, Terra WR. Substrate specificities of midgut β-glycosidases from insects of different orders. Comp Biochem Physiol B-Biochem Mol Biol 1998; 119: 219–225
  • Junge W, Wortmann W, Wilke B, Waldenstrom J, Kurrle-Weittenhiller A, Finke J, Klein G. Development and evaluation of assays for the determination of total and pancreatic amylase at 37°C according to the principle recommended by the IFCC. Clin Biochem 2001; 34: 607–615
  • Lorentz K. Routine α-amylase assay using protected 4-nitrophenyl-1,4-α-D-maltoheptaoside and a novel α-glucosidase. Clin Chem 2000; 46: 644–649
  • Rowland B, Purkayastha A, Monserrat C, Casart Y, Takiff H, McDonough KA. Fluorescence-based detection of lucZ reporter gene expression in intact and viable bacteria including Mycobacterium species. FEMS Microbiol Lett 1999; 179: 317–325
  • Skory CD, Freer SN, Bothast RJ. Properties of an intracellular β-glucosidase purified from the cellobiose-fermenting yeast Candida wickerhamii. Appl Microbiol Biotechnol 1996; 46: 353–359
  • Domingues L, Teixeira JA, Lima N. Rapid and sensitive detection of β-galactosidase-producing yeasts by using microtitre plate assay. Biotechnol Tech 1997; 11: 399–402
  • Ziser L, Setyawati I, Withers SG. Syntheses and testing of substrates and mechanism-based inactivators for xylanases. Carbohydr Res 1995; 274: 137–153
  • Laemmli UK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 1970; 227: 680–685
  • Armstrong JM, Myers DV, Verpoorte JA, Edsall JT. Purification and properties of human carbonic anhydrases. J Biol Chem 1966; 241: 5137–5149
  • Talalay P, Fishman WH, Huggins C. Chromogenic substrates. II. Phenolphthalein glucuronic acid as substrate for the assay of glucuronidase activity. J Biol Chem 1946; 166: 757–772
  • Lineweaver H, Burk D. The determination of enzyme dissociation constants. J Am Chem Soc 1934; 56: 658–666
  • Cornish-Bowden A, Eisenthal R. Statistic considerations in the estimation of enzyme kinetic parameters by the direct linear plot and other methods. Biochem J 1974; 139: 721–730
  • Eisenthal R, Cornish-Bowden A. The direct linear plot. A new graphical procedure for estimating enzyme kinetic parameters. Biochem J 1974; 139: 715–720
  • Grabowski GA, Osieckinewman K, Dinur T, Fabbro D, Legler G, Gatt S, Desnick RJ. Human acid β-glucuronidase - Use of conduritol-β-epoxide derivatives to investigate the catalytically active normal and Gaucher disease enzymes. J Biol Chem 1986; 261: 8263–8269
  • Witkowski A, Kindy MS, Daunert S, Bachas LG. Preparation of biotinylated β-galactosidase conjugates for competitive-binding assays by posttranslational modification of recombinant proteins. Anal Chem 1995; 67: 1301–1306
  • Brenzovich WE, Odell MD, Soares JP, Abelt CJ. Structure and properties of some cresolphthalein derivatives. Dyes and Pigments 2003; 59: 251–261

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.