2007 A4 HPLC Assay Anal Biochem

Embed Size (px)

Citation preview

  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    1/7

    ANALYTICAL

    BIOCHEMISTRY

    Analytical Biochemistry 363 (2007) 128134

    www.elsevier.com/locate/yabio

    0003-2697/$ - see front matter 2007 Elsevier Inc. All rights reserved.

    doi:10.1016/j.ab.2006.12.044

    Natural substrate assay for chitinases using high-performance liquidchromatography: A comparison with existing assays

    Inger-Mari Krokeide, Bjrnar Synstad, Sigrid Gseidnes, Svein J. Horn,Vincent G.H. Eijsink, Morten Srlie

    Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, P.O. Box 5003, N-1432 Aas, Norway

    Received 7 November 2006

    Available online 4 January 2007

    Abstract

    The determination of kinetic parameters of chitinases using natural substrates is diYcult due to low Km values, which require the use

    of low substrate concentrations that are hard to measure. Using the natural substrate (GlcNAc)4, we have developed an assay for the

    determination ofkcat and Kmvalues of chitinases. Product concentrations as low as 0.5 M were detected using normal-phase high-perfor-

    mance liquid chromatography (HPLC) with an amide 80 column (0.20 25 cm) using spectrophotometric detection at 210nm. By means

    of this assay, kcat and Kmvalues for chitinases A (ChiA) and B (ChiB) ofSerratia marcescens were found to be 331 s1 and 91 M and

    282 s1 and 4 2M, respectively. For ChiB, these values were compared to those found with commonly used substrates where the

    leaving group is a (nonnatural) chromophore, revealing considerable diVerences. For example, assays with 4-methylumbelliferyl-(Glc-

    NAc)2 yielded a kcat value of 18 2 s1 and a Km value of 306 M. For two ChiB mutants containing a Trp ! Ala mutation in the +1

    or +2 subsites, the natural substrate and the 4-methylumbelliferyl-(GlcNAc)2 assays yielded rather similar Km values (5-fold diVerence at

    most) but showed dramatic diVerences in kcat values (up to 90-fold). These results illustrate the risk of using artiWcial substrates for char-

    acterization of chitinases and, thus, show that the new HPLC-based assay is a valuable tool for future chitinase research. 2007 Elsevier Inc. All rights reserved.

    Keywords: Enzymatic assay; Chitinase; Natural substrate; HPLC

    Chitin, a -1,4-linked polymer of N-acetylglucosamine

    (GlcNAc), is an abundant biopolymer in nature. It is the

    most important nonplant structural biopolymer, occurring

    in, e.g., the exoskeletons of invertebrates, the cell walls of

    fungi, and the digestive tracts of insects. Chitin is easily

    derived from waste products such as shrimp shells. So far,

    chitin is primarily used as a source for chitosan, a partiallydeacetylated soluble form of chitin, and for glucosamine.

    Fragments of chitin or chitosan (chitooligosaccharides)

    may inhibit certain chitin-degrading enzymes, giving them

    potential as fungicides [1], insecticides [24], and antimala-

    rials [5,6]. Chitooligosaccharides are environmentally

    friendly because of their fast degradation in nature and,

    since chitin does not occur in humans, chitin metabolism is

    an interesting target area for development of drugs and

    pesticides.

    Chitin does not accumulate in nature because the poly-

    mer is eVectively degraded by diVerent chitinases belonging

    to the glycoside hydrolase enzyme families 18 and 19 [7].

    Serratia marcescens has an eYcient chitinolytic machinery

    and, when grown on chitin, three family 18 chitinases areexpressed: chitinases A (ChiA), B (ChiB), and C (ChiC) [8].

    ChiA and ChiB are processive chitinases that digest the

    chitin polymer in opposite directions producing mainly

    (GlcNAc)2, while ChiC is a nonprocessive endochitinase

    that hydrolyzes the polymer randomly, yielding longer chi-

    tooligosaccharides [9,10]. The S. marcescens chitinases have

    been characterized in several studies [811], using a variety

    of substrates.

    Kinetic analysis of chitinases is usually conducted with

    artiWcial substrates such as 4-methylumbelliferyl-(GlcNAc)2

    * Corresponding author. Fax: +47 64965901.

    E-mail address:[email protected] (M. Srlie).

    mailto:%[email protected]:%[email protected]:%[email protected]
  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    2/7

    Natural substrate assay for chitinases using HPLC / I.-M. Krokeide et al. / Anal. Biochem. 363 (2007) 128134 129

    ((GlcNAc)2-4MU), (GlcNAc)3-4MU, and p-nitrofenyl-

    (GlcNAc)2 ((GlcNAc)2-pNP). These substrates are not

    optimal because they present the enzyme with nonnatural

    leaving groups and because of substrate inhibition [12].

    Because of the length and the nature of the leaving groups,

    non-natural substrates are of very limited use when assess-

    ing the eVects of mutations meant to aVect natural sub-strate degradation. In addition, the leaving groups 4-MU

    and pNP are corresponding bases to weak acids with pKavalues of 7.8 [13] and 7.2 [14], respectively, while acid disso-

    ciation constants for sugars areV 12 [15]. At high pH (Glc-

    NAc)2-4MU and (GlcNAc)2-pNP yield negatively charged

    leaving groups while this will not be the case for a sugar

    leaving group. Kinetic characterization with natural sub-

    strates is diYcult because (1) low Km values require detec-

    tion of low concentrations of substrate and product, (2)

    most longer substrates contain more than one hydrolysable

    glycosidic bond, (3) degradation products longer than

    dimers are also substrates, and (4) some chitinases degrade

    longer substrates processively. With respect to the latter

    two problems, (GlcNAc)4 is an exception since most chitin-

    ases convert this compound exclusively to two dimers,

    which are not degraded any further.

    We present a chitinase assay based on the use of the natu-

    ral substrate (GlcNAc)4 and the use of a sensitive HPLC

    setup to monitor substrate and product concentrations. We

    have compared the natural substrate assay with existing

    assays using wild-type ChiA and ChiB from S. marcescens

    and two engineered variants of ChiB. The engineered enzymes

    are ChiB-W97A and ChiB-W220A. Trp97 is located in the +1

    subsite of the active site of ChiB while Trp220 is located in the

    +2 subsite [16,17]. Mutation of these residues will thus aVectthe enzymes interaction with the leaving group.

    Materials and methods

    Chemicals

    Tetra-N-acetylchitotetraose, 4-methylumbelliferyl-di-N-

    acetylchitobiose, 4-methylumbelliferyl-tri-N-acetylchitotrii-

    ose, para-nitrophenyl-di-N-acetylchitobiose, and acetonitrile

    were purchased from SigmaAldrich.

    Production and puriWcation of chitinases

    ChiA [18], ChiB [19], and mutants of ChiB were puriWed

    from periplasmatic extracts of the producer strains by

    hydrophobic interaction chromatography, as described

    previously [12]. Enzyme purity was veriWed using SDS/

    PAGE and was above 95% in all cases. Protein concentra-

    tions were determined using the Bradford assay kit

    provided by Bio-Rad (Hercules, CA, USA).

    Chromatography of chitooligosaccharides

    Mixtures of (GlcNAc)4 and (GlcNAc)2 were separated

    by normal-phase HPLC using a Tosoh TSK Amide 80

    column (0.2025 cm) with an amide 80 guard column. The

    sample size was 50L and the chitooligosaccharides were

    eluted isocratically at 0.25mL/min with 70% (v/v) acetoni-

    trile at room temperature. The chitooligosaccharides were

    monitored by measuring absorbance at 210nm and the

    (GlcNAc)4 concentrations were quantiWed by measuring

    peak areas and by comparing these to those of standardsamples with known concentrations of (GlcNAc)4. Using

    these standard samples, it was established that there was a

    linear correlation between the peak area and the analyzed

    (GlcNAc)4 concentration within the concentration range

    0.5300M used in this study.

    (GlcNAc)4 assay

    Reactions were started by adding 0.5nM ChiA or ChiB

    or 1.0 nM ChiB-W220A or 2.0nM ChiB-W97A (Wnal con-

    centrations) to 1-mL solutions containing 100, 80, 60, 40,

    20, 10, 5, or 2M (for ChiA and ChiB), 500, 400, 300, 200,

    100, 50, or 25 M (for ChiBW220A), and 1000, 600, 400,

    200, 100, 50, or 25 M (for ChiB-W97A) (GlcNAc)4 in

    20 mM NaAc buVer, pH 6.1, and 0.1 mg/mL bovine serum

    albumin (Wnal concentrations). After enzyme addition, 50-

    L aliquots were transferred to a HPLC vial containing

    150L of acetonitrile at appropriate time points. Reaction

    conditions and timing were such that the (GlcNAc)4 con-

    centration in the sample would not go below 80% of the

    starting concentration. An aliquot taken before enzyme

    addition was used as the tD 0 sample. The slopes of the

    plots of substrate concentrations vs time were taken as the

    hydrolysis rate. Then the hydrolysis rates were plotted vs

    the substrate concentration in a MichaelisMenten plotand the experimental data were Wtted to the Michaelis

    Menten equation using nonlinear Wtting in Origin 7. Alter-

    natively, substrate concentrations divided by the hydrolysis

    rates were plotted vs substrate concentrations in a Hanes

    plot using a standard spreadsheet program to obtain kcatand Km values. The derived values using the two diVerent

    approaches were the same within experimental errors.

    Assays with nonnatural substrates

    The kinetic parameters of ChiB variants for the (Glc-

    NAc)2-4MU substrate at pH 6.1 were determined as thor-oughly described elsewhere [12,20] with an enzyme

    concentration of 0.2nM and substrate concentrations in the

    5 to 50-M range. The kinetic parameters of wild-type

    ChiB toward (GlcNAc)2-pNP were determined in the same

    way, except that the substrate concentration range was

    adapted to the much higher Km for this substrate. ChiB

    converts the (GlcNAc)3-4MU substrate exclusively to (Glc-

    NAc)2 and (nondetectable) GlcNAc-4MU. The kinetic

    parameters of wild-type ChiB toward the (GlcNAc)3-4MU

    substrate were determined using a two-step assay based on

    detection of substrate depletion, a ChiB concentration of

    0.2 nM, and substrate concentrations in the 1 to 10-M

    range [21]. In short, reactions were conducted as usual but,

  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    3/7

    130 Natural substrate assay for chitinases using HPLC / I.-M. Krokeide et al. / Anal. Biochem. 363 (2007) 128134

    before adding the 0.2M Na2CO3 stop solution, the reaction

    mixtures were incubated for less than 1 min with a 300-fold

    excess of ChiA to convert all remaining substrate to

    (detectable) free 4MU. In all cases, the substrate concentra-

    tions divided by the hydrolysis rates were plotted vs sub-

    strate concentration in Hanes plots using a standard

    spreadsheet program to obtain kcat and Km values.

    Results

    (GlcNAc)4 as substrate

    (GlcNAc)4 productively binds in subsites 2 to +2 of

    ChiA and ChiB of S. marcescens, yielding two (GlcNAc)2molecules as products that are not subject to further hydro-

    lysis [8,12,21]. Longer substrates have several productive

    binding modes and yield products that in turn are sub-

    strates for the enzymes, complicating kinetic analysis of the

    overall reaction. Productive binding of (GlcNAc)3 yields

    only products that are not further hydrolyzed, (GlcNAc)2and GlcNAc. However, this substrate is less suitable than

    (GlcNAc)4 because it occupies fewer subsites and because

    there are two productive binding modes, at least in some

    chitinases such as ChiA (binding in 2 to +1 and in 1

    to +2) [21]).

    HPLC analysis of chitooligosaccharides

    Normal-phase HPLC-based quantitative analysis of sac-

    charides with an amide 80 column has been used previously

    [2124] but, so far, the use of this method for determination

    of kinetic parameters (implying measurement of very lowconcentrations of sugar) has not been reported. In the pres-

    ent study, the necessary increase in sensitivity was achieved

    through a combination of adjustments. First, we used a col-

    umn with a considerably reduced diameter (0.20cm vs

    0.48 cm used in previous studies). A smaller column diame-

    ter yields a smaller degree of sample dilution compared to a

    larger column diameter and thus results in sharper peaks.

    Second, injection volumes were 50 L. Since water elutes

    the chitooligosaccharides, the samples were diluted to 75%

    (v/v) acetonitrile before application to allow for retardation

    on the column of the chitiooligosaccharides before elution

    with 70% (v/v) acetonitrile. Together these adjustments per-mitted detection of (GlcNAc)4 concentrations down to

    0.5M. Fig. 1 shows the HPLC chromatograms resulting

    from enzymatic hydrolysis of (GlcNAc)4 by ChiB.

    Determination of kcat, Km, and eYciency constants (kcat/Km)

    for wild-type ChiB with diVerent substrates

    Enzymatic hydrolysis of (GlcNAc)4 by ChiB follows

    MichaelisMenten kinetics as depicted by Fig. 2A. Kinetic

    data for hydrolysis of (GlcNAc)4 and three diVerent artiW-

    cial substrates are shown in Table 1. For (GlcNAc)4 hydro-

    lysis, the nonlinear Wt of theoretical data to experimental

    data yielded kcat and Km values of 282 s1 and 4 2M,

    respectively. These values diVer considerably from values

    obtained with the often-used substrate (GlcNAc)2-4MU,

    which are 18 2 s1 and 316M, respectively [20]. Even

    larger diVerences were observed with the (GlcNAc)2-pNP

    substrate which yielded kcat and Km values of 1.40.5s-1

    and 18135M, respectively. For (GlcNAc)3-4MU (which

    is converted to (GlcNAc)2 and GlcNAc-4MU), the kcatvalue is 574 s1 and the Km value is 71M and both are

    about two times larger than those for (GlcNAc)4.

    The diVerences between the substrates are most apparent

    when using eYciency constants (kcat/Km; Table 1). ChiB has

    a 700 times greater eYciency toward (GlcNAc)4 compared

    to (GlcNAc)2-pNP and a 12 times higher eYciency

    compared to (GlcNAc)2-4MU. Results obtained with the

    (GlcNAc)3-4MU substrate were similar to those obtained

    with (GlcNAc)4.

    Determination of kinetic parameters for ChiA with

    (GlcNAc)4

    The (GlcNAc)2-4MU susbtrate is often used because its

    product is easy to detect at low concentrations and because

    it tends to display seemingly natural Km values, i.e., in the

    lower micromolar range. However, for some enzymes with

    extended substrate binding clefts this short substrate may

    be suboptimal because of the occurrence of multiple bind-

    ing modes and cooperativity between these. This is for

    example observed for ChiA [12]. With the (GlcNAc)4 sub-

    strate, ChiA displayed normal MichaelisMenten kinetics

    (Fig. 2B), yielding kcat and Km values of 331 s1 and

    91M, respectively (Table 2).

    Fig. 1. HPLC analysis of (GlcNAc)4 hydrolysis by ChiB. The starting con-

    centration of (GlcNAc)4 was 40 M, and chromatograms obtained after

    hydrolysis of 0% (bottom), approximately 10% (middle), and 20% (top) of

    the substrate are shown. Both the tetramer and the dimer peak are split

    into two peaks due to the anomer equilibrium of the chitooligosaccha-

    rides. The anomers (approximately 60%) elute before the anomers

    (approximately 40%; see, e.g., [21]). Due to the small diameter of the col-

    umn and the relatively large sample volume, the anomeric forms are only

    partly separated. The Wrst 8 min of the chromatograms were omitted for

    clarity.

    8 10 12 14 16 18 20

    0

    5

    10

    15

    20

    25

    Absorbance/mV

    Time /min

  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    4/7

    Natural substrate assay for chitinases using HPLC / I.-M. Krokeide et al. / Anal. Biochem. 363 (2007) 128134 131

    Determination of kcat and Km values for engineered chitinases

    using (GlcNAc)4 and (GlcNAc)2-4MU as substrates

    Activity assays based on natural substrates are particu-larly important for the analysis of the properties of engi-

    neered enzymes carrying site-directed mutations near the

    catalytic center. To analyze and illustrate this, we have char-

    acterized two ChiB variants with mutations in subsite +1

    (W97A) and +2 (W220A) (Fig. 3) with both (GlcNAc)4 and

    (GlcNAc)2-4MU (Table 2). When (GlcNAc)4 was used, kcatand Km values of 126 2 s

    1 and 807 2M were obtained

    for ChiB-W97A, while the same values were 81 s1 and

    17557M for the (GlcNAc)2-4MU substrate. For ChiB-

    W220A, the kcat and Km values were 451 s1 and

    71 1M and 0.50.1s1 and 824M when using (Glc-

    NAc)4 and (GlcNAc)2-4MU, respectively. Interestingly,while the diVerence between the substrates is modest with

    respect to Km values, it is as large as 90-fold with respect to

    kcat values.

    Discussion

    Comparison of the various substrates

    Up to now, (GlcNAc)2-4MU, which produces the

    Xuorescing 4MU leaving group upon hydrolysis, has been

    the most used substrate in chitinase assays [12,20,22,25].

    Obvious advantages of the use of (GlcNAc)2-4MU are the

    time window and simplicity of the assay and its sensitivity.

    Product formation is readily and quickly detected using

    Xuorescence spectroscopy. There are also several and seri-

    ous disadvantages such as substrate inhibition, a nonnatu-

    ral leaving group, and nonlinear kinetics, [12,22,26]. Analternative for (GlcNAc)2-4MU is (GlcNAc)2-pNP, which

    yields the yellow chromophore pNP. This substrate has the

    same disadvantages as (GlcNAc)2-4MU. The data ofTable 1

    show that the kinetic parameters determined with

    (GlcNAc)2-pNP are very diVerent from the presumably

    more realistic parameters obtained with (GlcNAc)4.

    Another alternative is (GlcNAc)3-4MU, which may be

    converted either to (GlcNAc)2 and GlcNAc-4MU (as does

    ChiB) or to (GlcNAc)3 and 4MU (as does ChiA) [12]. In the

    latter case, product formation can readily be determined,

    but the method has most of the disadvantages described

    above for (GlcNAc)2-4MU. For ChiB, we have been able to

    develop a method for measuring the conversion of (Glc-

    NAc)3-4MU to (GlcNAc)2 and GlcNAc-4MU under con-

    ditions that permit determination of kinetic parameters

    [21]. The method is based on measuring product disappear-

    ance, which is achieved by converting remaining product

    quantitatively to 4MU through a short incubation with a

    large excess of ChiA. This assay has the advantage that the

    +1 subsite is occupied by a natural leaving group (i.e., a

    sugar unit). Not unexpectedly, the results obtained with

    (GlcNAc)3-4MU for wild-type ChiB resembled those

    obtained with (GlcNAc)4. A serious disadvantage of this

    assay is the need to purify two enzymes, especially large

    amounts of ChiA, which is time consuming, and it works

    Fig. 2. MichaelisMenten plots from HPLC analyses of (GlcNAc)4 hydrolysis with ChiB (A), ChiA (B), and the engineered enzymes ChiB-W220A (C) and

    ChiB-W97A (D) at pH 6.1, 37 C. Solid symbols are experimentally determined data for the rate of substrate disappearance vs substrate concentrations

    and the solid lines are the best nonlinear Wt using the MichaelisMenten equation.

    0 20 40 60 80 100

    0,000

    0,005

    0,010

    0,015

    0,020

    R

    ate/(M/s)

    (GlcNAc)4/M

    0 100 200 300 400 500

    0,00

    0,01

    0,02

    0,03

    0,04

    (GlcNAc)4/M

    R

    ate/(M/s)

    0 20 40 60 80 100

    0,000

    0,005

    0,010

    0,015

    0,020

    Rate/(M/s)

    (GlcNAc)4/M

    0 200 400 600 800 1000 1200

    0.00

    0.05

    0.10

    0.15

    Rate/(M/s)

    (GlcNAc)4/M

    A C

    DB

  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    5/7

    132 Natural substrate assay for chitinases using HPLC / I.-M. Krokeide et al. / Anal. Biochem. 363 (2007) 128134

    only for enzymes that exclusively convert (GlcNAc)3-4MU

    to (GlcNAc)2 and GlcNAc-4MU.

    Although the rate-limiting step in the reaction mecha-

    nism of family 18 chitinases is not known [17,20,27], it is

    clear that a change in chemical properties of the leaving

    group may aVect catalytic eYciency with regard to both the

    pKa and the ability to interact with the enzyme. At pH 6.1,

    about 2% of 4-methylumbelliferol and about 7% ofpara-

    nitrophenol will be negatively charged while virtually all of

    the (GlcNAc)2 and GlcNAc-4MU leaving groups will be

    protonated and free of charge. It is interesting to note that

    the substrate yielding the by far lowest kcat value ((Glc-

    NAc)2-pNP) has the most acidic and the smallest leaving

    group. ChiB is more eVective toward (GlcNAc)2-4MU

    whose leaving group is slightly less acidic and considerably

    larger. Several studies have shown that enzymesubstrate

    interactions in subsites +1 and +2 are important for

    catalytic eYciency [17,22,28,29]. The present data do not

    permit discrimination between the roles of the acidity and

    Ta le 1

    kcat and Km of ChiB from Serratia marcescens for substrates with diVerent leaving groups, at 37 C, pH 6.1

    a Ref[21].b Ref[20].

    Leaving group (RH) kcat (s1) Km (M) kcat/Km (s

    1 M1)

    Di-N-acetylchitobiose

    28 2 4 2 7

    4-Methylumbelliferyl-N-acetylglucosamine

    57 4a 7 1a 8

    4-Methylumbelliferol

    18 2b 31 6b 0.6

    ara-Nitrophenol

    1.4 0.5 181 35 0.01

    O

    ROH

    OH

    NH

    O

    OOH O

    OH

    OH

    NH

    O

    O

    OHOH

    OH

    NH

    O

    OOH O

    OH

    OH

    NH

    O

    + RHH2O

    O

    OH

    OH

    NH

    O

    OO

    OH

    OH

    NH

    O

    OHOH

    O

    OO

    OH

    OH

    NH

    O

    OH

    O

    O OOH

    OH

    NO

    O

    Ta le 2

    Kinetic parameters for engineered and natural chitinases from Serratia

    marcescens at 37C, pH 6.1, using (GlcNAc)4 and 4MU-(GlcNAc)2 as

    substrates

    a (s1).b (M).c (s1M1).d The combination of sigmoidal behavior and substrate inhibition

    precludes determination of kinetic parameters [12].

    Chitinase (GlcNAc)4 (GlcNAc)2-4MU

    kcata Km

    b kcat/Kmc kcat

    a Kmb kcat/Km

    c

    ChiB 28 2 4 2 7 18 2 31 6 0.6

    ChiA 33 1 9 1 4 n.d.d n.d.d n.d.d

    ChiB-W97A 126 4 807 40 0.2 8 1 175 57 0.03

    ChiB-W220A 45 2 71 3 0.6 0.5 0.1 82 4 0.01

  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    6/7

    Natural substrate assay for chitinases using HPLC / I.-M. Krokeide et al. / Anal. Biochem. 363 (2007) 128134 133

    the interaction potential of the leaving group. The data in

    Table 1 do suggest though that both the occupancy of the +1

    and +2 subsites and a high pKa of the leaving group are

    beneWcial for catalytic eYciency because the longer, sugar-

    like substrates yielded lower Km and higher kcat values.

    The present data show that the use of (GlcNAc)4 to cir-

    cumvent the problems connected to other substrates is fully

    feasible. Both ChiA and ChiB showed straightforward

    MichaelisMenten kinetics without substrate inhibition

    and the range of accessible substrate concentrations was

    such that kinetic parameters could be determined with rea-sonable accuracy. The results show that ChiA and ChiB

    have similar activities toward soluble substrates, in line

    with the rather similar architectures of their active sites and

    substrate-binding grooves [11,16,17,30]).

    Analysis of mutants

    The W97A and W220A mutants displayed large

    increases in Km with both substrates, as might be expected

    upon mutating important interaction partners in subsites

    that are known to make a dominant positive contribution

    to ligand binding. The increase in Km was larger for the nat-ural substrate (GlcNAc)4, presumably because the wild-

    type subsites are optimized for sugar binding rather than

    binding of the 4MU group. Not unexpectedly, mutation of

    Trp220, which primarily aVects subsite +2, had only a mod-

    est eVect on the Km for (GlcNAc)2-4MU.

    Mutational eVects on kcat showed a dramatic dependence

    on the substrate used (Table 2). With the natural substrate,

    both mutations lead to an increase in kcat (4.5 and 1.6 times

    higher for ChiB-W97A and W220A, respectively) whereas

    the kcat is reduced from 18 to 8s1 for ChiB-W97A and to

    0.5s1 for ChiB-W220A with (GlcNAc)2-4MU. For the

    ChiB-W220A mutant, the two substrates yield a 90-fold

    diVerence in kcat and a 60-fold diVerence in the eYciency

    constant kcat/Km. Apart from illustrating the diVerences

    between the substrates, these observations also lead to ques-

    tions concerning the mechanism and rate-limiting step of

    catalysis. For example, the dramatic eVect of the ChiB-

    W220A mutation on the kcat obtained with (GlcNAc)2-

    4MU must mean that Trp220 is involved in catalysis, even in

    the case of short substrates. It is not known how the 4MUgroup binds to the enzyme, but it is clear that the group is

    large enough (i.e., larger than a single sugar) to interact with

    parts of the +2 subsite, including Trp220. Another puzzling

    issue concerns the fact that mutation of the two tryptophans

    increased the kcat for the natural substrate. Interestingly,

    Watanabe et al. [31] found that mutation of analogous try-

    ptophans in chitinase A1 from Bacillus circulans led to

    increased speciWc activity toward chitopentaose. Further

    kinetic and mutational studies are necessary to Wnd possible

    explanations for these observations [32].

    In conclusion, the results obtained with wild-type ChiB

    and, particularly, with the two mutants clearly show that

    the use of easy-to-handle artiWcial substrates for detailed

    characterization of family 18 chitinases may lead to wrong

    conclusions. Thus, such characterization should be based

    on a natural substrate assay, such as the one described here.

    Acknowledgments

    We are grateful for the help of Dr. Gustav Vaaje-

    Kolstad for the making of Fig. 3. Part of this work was

    funded by The Norwegian Research Council, Grants

    140497 and 140440.

    References

    [1] H. Izumida, M. Nishijima, T. Takadera, A.M. Nomoto, H. Sano, The

    eVect of chitinase inhibitors, cyclo(Arg-Pro) against cell separation of

    Saccharomyces cerevisiae and the morphological change of Candida

    albicans, J. Antibiot.(Tokyo) 49 (1996) 829831.

    [2] E. Cohen, Chitin synthesis and degradation as targets for pesticide

    action, Arch. Insect Biochem. Physiol. 22 (1993) 245261.

    [3] S. Sakuda, A. Isogai, S. Matsumoto, A. Suzuki, Search for microbial

    insect growth regulators. II. Allosamidin, a novel insect chitinase

    inhibitor, J. Antibiot. (Tokyo) 40 (1987) 296300.

    [4] K. Shiomi, N. Arai, Y. Iwai, A. Turberg, H. Kolbl, S. Mura, Structure

    of argiWn, a new chitinase inhibitor produced by Gliocladium sp, Tet-

    rahedron Lett. 41 (2000) 21412143.

    [5] J.M. Vinetz, S.K. Dave, C.A. Specht, K.A. Brameld, B. Xu, R. Hay-ward, D.A. Fidock, The chitinase PfCHT1 from the human malaria

    parasite Plasmodium falciparum lacks proenzyme and chitin-binding

    domains and displays unique substrate preferences, Proc. Natl. Acad.

    Sci. USA 96 (1999) 1406114066.

    [6] J.M. Vinetz, J.G. Valenzuela, C.A. Specht, L. Aravind, R.C. Langer,

    J.M.C. Ribeiro, D.C. Kaslow, Chitinases of the Avian Malaria Para-

    site Plasmodium gallinaceum, a Class of Enzymes Necessary for Para-

    site Invasion of the Mosquito Midgut, J. Biol. Chem. 275 (2000)

    1033110341.

    [7] B. Henrissat, G. Davies, Structural and sequence-based classiWca-

    tion of glycoside hydrolases, Curr. Opin. Struct. Biol. 7 (1997)

    637644.

    [8] K. Suzuki, N. Sugawara, M. Suzuki, T. Uchiyama, F. Katouno, N.

    Nikaidou, T. Watanabe, Chitinases A, B, and C1 ofSerratia marces-

    cens 2170 produced by recombinant Escherichia coli: enzymatic prop-

    Fig. 3. Crystal structure of the active site of ChiB with (GlcNAc)5 bound

    [17]. Trp97 stacks with the sugar moiety in subsite +1. Similarly, Trp220

    stacks with the sugar moiety in subsite +2. Note that the 1 sugar has a

    non-chair conformation that resembles a sofa conformation [27], the

    occurrence of which is crucial during the catalytic cycle [17,27]. Clearly,

    mutations that aVect substrate binding may also aVect this crucial struc-

    tural deformation of the sugar. Note. The complex was determined with

    an inactive E144Q mutant of ChiB. For illustration purposes, the picture

    shows a glutamate.

  • 8/6/2019 2007 A4 HPLC Assay Anal Biochem

    7/7

    134 Natural substrate assay for chitinases using HPLC / I.-M. Krokeide et al. / Anal. Biochem. 363 (2007) 128134

    erties and synergism on chitin degradation, Biosci. Biotechnol.

    Biochem. 66 (2002) 10751083.

    [9] S.J. Horn, A. Sorbotten, B. Synstad, P. Sikorski, M. Sorlie, K.M.

    Varum, V.G.H. Eijsink, Endo/exo mechanism and processivity of

    family 18 chitinases produced by Serratia marcescens, FEBS J. 273

    (2006) 491503.

    [10] E.L. Hult, F. Katouno, T. Uchiyama, T. Watanabe, J. Sugiyama,

    Molecular directionality in crystalline beta-chitin: hydrolysis by

    chitinases A and B from Serratia marcescens 2170, Biochem. J. 388

    (2005) 851856.

    [11] N.N. Aronson, B.A. Halloran, M.F. Alexyev, L. Amable, J.D. Madura,

    L. Pasupulati, C. Worth, P. Van Roey, Family 18 chitinase-oligosaccha-

    ride substrate interaction: subsite preference and anomer selectivity of

    Serratia marcescens chitinase A, Biochem. J. 376 (2003) 8795.

    [12] M.B. Brurberg, I.F. Nes, V.G.H. Eijsink, Comparative studies of

    chitinases A and B from Serratia marcescens, Microbiology 142

    (1996) 15811589.

    [13] W.C. Sun, K.R. Gee, R.P. Haugland, Synthesis of novel Xuorinated

    coumarins: Excellent UV-light excitable Xuorescent dyes, Bioorg.

    Med. Chem. Lett. 8 (1998) 31073110.

    [14] Pawel. Wiczling, Michal J. Markuszewski, Roman. Kaliszan, Deter-

    mination of pKa by pH Gradient Reversed-Phase HPLC, Anal.

    Chem. 76 (11) (2004) 30693077.[15] B. Capon, W.G. Overend, Constitution and physicochemical proper-

    ties of carbohydrates, Adv. Carb. Chem. 15 (1960) 1151.

    [16] D.M.F. van Aalten, B. Synstad, M.B. Brurberg, E. Hough, B.W. Riise,

    V.G.H. Eijsink, R.K. Wierenga, Structure of a two-domain chitotrios-

    idase from Serratia marcescens at 1.9-angstrom resolution, Proc. Natl.

    Acad. Sci. USA 97 (2000) 58425847.

    [17] D.M.F. Van Aalten, D. Komander, B. Synstad, S. Gseidnes, M.G.

    Peter, V.G.H. Eijsink, Structural insights into the catalytic mechanism

    of a family 18 exo-chitinase, Proc. Natl. Acad. Sci. USA 98 (2001)

    89798984.

    [18] M.B. Brurberg, V.G.H. Eijsink, I.F. Nes, Characterization of a chitinase

    gene (chiA) from Serratia marcescens BJL200 and one-step puriWcation

    of the gene product, FEMS Microbiol. Lett. 124 (1994) 399404.

    [19] M.B. Brurberg, V.G.H. Eijsink, A.J. Haandrikman, G. Venema, I.F.

    Nes, Chitinase B from Serratia marcescens BJL200 is exported to theperiplasm without processing, Microbiology 141 (1995) 123131.

    [20] B. Synstad, S. Gseidnes, D.M.F. van Aalten, G. Vriend, J.E. Nielsen,

    V.G.H. Eijsink, Mutational and computational analysis of the role of

    conserved residues in the active site of a family 18 chitinase, Eur. J.

    Biochem. 271 (2004) 253262.

    [21] S.J. Horn, M. Srlie, G. Vaaje-Kolstad, A.L. Norberg, B. Synstad,

    K.M. Vrum, V.G.H. Eijsink, Comparative studies of chitinases A, B

    and C from Serratia marcescens, Biocatal. Biotransfor. 24 (2006) 39

    53.

    [22] T. Fukamizo, C. Sasaki, E. Schelp, K. Bortone, J.D. Robertus, Kinetic

    properties of chitinase-1 from the fungal pathogen Coccidioides imm-

    itis, Biochemistry 40 (2001) 24482454.

    [23] T. Toyooka, A. Kuze, Determination of saccharides labelled with a

    Xuorescent reagent, DBD-ProCZ, by liquid chromatography, Bio-

    med. Chromatogr. 11 (1997) 132136.

    [24] S. Chiye, I. Yoshifumi, T. Hideki, K. Satoru, F. Tamo, Family 19 chiti-

    nase from rice (Oryza sativa L.): substrate-binding subsites demon-

    strated by kinetic and molecular modeling studies, Plant Mol. Biol.

    V52 (2003) 4352.

    [25] Y. Honda, S. Tanimori, M. Kirihata, S. Kaneko, K. Tokuyasu, M.

    Hashimoto, T. Watanabe, T. Fukamizo, Kinetic analysis of the reac-

    tion catalyzed by chitinase A1 from Bacillus circulans WL-12 toward

    the novel substrates, partially N-deacetylated 4-methylumbelliferyl

    chitobiosides, FEBS Lett. 476 (2000) 194197.

    [26] C. Malet, A. Planas, Mechanism of Bacillus 1,3-1,4-beta-D-glucan 4-

    glucanohydrolases: kinetics and pH studies with 4-methylumbelliferyl

    beta-D-glucan oligosaccharides, Biochemistry 36 (1997) 1383813848.

    [27] I. Tews, A.C.T. vanScheltinga, A. Perrakis, K.S. Wilson, B.W. Dijk-stra, Substrate-assisted catalysis uniWes two families of chitinolytic

    enzymes, J. Am. Chem. Soc. 119 (1997) 79547959.

    [28] Y. Honda, T. Fukamizo, Substrate binding subsites of chitinase from

    barley seeds and lysozyme from goose egg white, Biochim. Biophys.

    Acta 1388 (1998) 5365.

    [29] C. Sasaki, Y. Itoh, H. Takehara, S. Kuhara, T. Fukamizo, Family 19

    chitinase from rice (Oryza sativa L.): substrate-binding subsites dem-

    onstrated by kinetic and molecular modeling studies, Plant Mol. Biol.

    52 (2003) 4352.

    [30] A. Perrakis, I. Tews, Z. Dauter, A.B. Oppenheim, I. Chet, K.S. Wilson,

    C.E. Vorgias, Crystal structure of a bacterial chitinase at 2.3 A resolu-

    tion, Structure 2 (1994) 11691180.

    [31] T. Watanabe, Y. Ariga, U. Sato, T. Toratani, M. Hashimoto, N. Nikai-

    dou, Y. Kezuka, T. Nonaka, J. Sugiyama, Aromatic residues within the

    substrate-binding cleft ofBacillus circulans chitinase A1 are essential forhydrolysis of crystalline chitin, Biochem. J. 376 (2003) 237244.

    [32] S.J. Horn, P. Sikorski, J.B. Cederkvist, G. Vaaje-Kolstad, M. Srlie,

    B. Synstad, G. Vriend, K.M. Vrum, V.G.H. Eijsink, Costs and bene-

    Wts of processivity in enzymatic degradation of recalcitrant polysac-

    charides, Proc. Natl. Acad. Sci. USA 103 (2006) 1808918094.