no, /ul/67531/metadc330887/m2/1/high_re… · the conversion of phenylglyoxal to mandelic acid,...

121
3>7<? A ; d; o No, /U PURIFICATION AND STUDIES OF MAMMALIAN GLYOXALASE ENZYMES DISSERTATION Presented to the Graduate Council of the North Texas State University in Partial Fulfillment of the Requirements For the Degree of Doctor of Philosophy By Bedii Oray, B. S. Denton, Texas December, 1980

Upload: others

Post on 23-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • 3>7

  • -u /,

    Gray, Bedii, Purification and Studies of Mammalian

    Glyoxalase Enzymes. Doctor of Philosophy (Chemistry),

    December, 1980, 110 pp., 5 tables, references, 96 titles.

    The glyoxalase system, which has been known since 1913,

    is widely distributed in nature. The system consists of two

    enzymes, glyoxalase I and glyoxalase II. The reactions

    catalyzed by the two enzymes are:

    glyoxalase I

    Methylglyoxal + glutathione

    glyoxalase II

    S-lactoylglutathione — ^

    H 2 °

    B-lactic acid + glutathione

    Methylglyoxal is very unstable and undergoes oxidation

    and polymerization reactions. One of the purposes of this

    study was to find a simple, convenient and reproducible

    method of methylglyoxal preparation. Another objective was

    the purification of both glyoxalase enzymes employing

    affinity chromatography as a major step. The purified

    enzymes were to be characterized by chemical, physical and

    kinetic properties as an approach to the understanding of

    the biological function of the system.

    The purification procedures were designed to minimize

    any alteration of enzyme proteins and to give high recovery

  • of the activities. The purification of glyoxalase I was

    accomplished by the use of both hydrophobic and affinity

    chromatography. The homogeneous enzyme has a specific

    activity of 944 units per mg protein. The enzyme has a

    molecular weight of 43,000 daltons; it is a dimer and is

    apparently composed of identical subunits of molecular

    weight approximating 21,500 daltons.

    The purification of glyoxalase II was accomplished by a

    rapid, two-step affinity chromatographic scheme. The homo-

    geneous enzyme exhibited a specific activity of 920 units

    per mg protein, has a molecular weight of approximately

    29,500 daltons and appears to be a monomer.

    Inhibition studies revealed that nucleotides, nucleo-

    sides, structurally related compounds, and also some

    microtubule poisons are potent, cooperative inhibitors of

    both glyoxalase enzymes. From the studies of nucleotides

    and related compounds, it was concluded that aromatic

    compounds which contain heterocyclic nitrogen and/or have

    amino groups on the aromatic ring, are effective inhibitors.

    Kinetic studies indicated that there is a common

    binding site on glyoxalase I for glutathione, S-octyl-

    glutathione, methylglyoxal, colchicine and the hemimercaptal

    of methylglyoxal and glutathione. Colchicine also seems to

    become a better inhibitor in the presence of increasing

    concentrations of free glutathione or free methylglyoxal.

  • The substrate of glyoxalase I, the hemimercaptal of methyl-

    glyoxal and glutathione, is a competitive inhibitor of

    glyoxalase II.

  • Copyright by

    Bedii Oray

    1980

    1 1 1

  • PREFACE

    The author expresses his gratitude to Mr. Michael W.

    Kellum for his help in the methylglyoxal synthesis pro-

    ject and to Mrs. Vana L. Smith, Ms. Jana Jordan and

    Mrs. Patricia Lambert for their technical assistance in

    this research project.

    The financial support of this investigation by the

    Robert A. Welch Foundation of Texas, No. B-133, and North

    Texas State University Faculty Research Fund is gratefully

    acknowledged.

    Bedii Oray

    December, 1980

    IV

  • TABLE OF CONTENTS

    Page

    LIST OF TABLES v i

    LIST OF ILLUSTRATIONS i • • • • • • • • • • v n

    Chapter

    I. INTRODUCTION x

    II. EXPERIMENTAL PROCEDURE n

    General Methylglyoxal Synthesis Routine Glyoxalase I Assays Routine Glyoxalase II Assays Determination of Protein Concentration Column Material Preparation Purification of Glyoxalase I Purification of Glyoxalase II Concentration Procedures Molecular Weight Determination of Glyoxalase I

    by Sedimentation Equilibrium Molecular Weight Determinations of Glyoxalase

    Enzymes by Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis

    Polyacrylamide Gel Electrophoresis Isoelectric Focusing Kinetic Studies

    III. RESULTS AND DISCUSSION 22

    Methylglyoxal Synthesis Purification of Glyoxalase I Purification of Glyoxalase II Molecular Weight Determinations Inhibition of Glyoxalase Enzymes by Nucleo-

    tides, Nucleosides and a Series of Structurally Related Compounds

    Inhibition of Glyoxalase Enzymes by Micro-tubule Poisons

    Kinetics

    IV. REFERENCES

  • LIST OF TABLES

    T a b l e Page

    I. Hydrophobic Chromatography of Glyoxalase I . . . 34

    II. Purification of Glyoxalase I 37

    III. Purification of Glyoxalase II 44

    IV. I 5 0 Values (pM) for Some Glyoxalase Inhibitors . 68

    V. Effect of Microtubule Poisons on Various Enzymes. 77

    VI

  • LIST OF ILLUSTRATIONS

    F i 9 u r e Page

    1. (A) Effect of Temperature on Hydrolysis of Pyruvaldehyde Dimethyl Acetal

    (B) Rate of Hydrolysis of Pyruvaldehyde Dimethyl Acetal 25

    2. Effect of Acid Concentration on Hydrolysis of Pyruvaldehyde Dimethyl Acetal 27

    3. Hydrolysis of Varying Levels of Pyruvaldehyde Dimethyl Acetal 29

    4. Glyoxalase I Kinetic Study with Methylglyoxal Prepared by the Hydrolysis of Pyruvalde-hyde Dimethyl Acetal 32

    5. Ethylamine—Sepharose Hydrophobic Chromatography of Swiss Mouse Liver Glyoxalase I 39

    6. S-Octylglutathione-Sepharose Affinity Chromatog-raphy of Swiss Mouse Liver Glyoxalase I . . 41

    7. First Oxidized Glutathione—Sepharose Affinity Chromatography of Swiss Mouse Liver Glyoxalase II 4 6

    8• Second Oxidized Glutathione—Sepharose Affinity Chromatography of Swiss Mouse Liver Glyoxalase II 49

    9. Sedimentation Equilibrium Ultracentrifugation of Glyoxalase I 5 2

    10. Molecular Weight Determination of Glyoxalase I by Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis 55

    11. Molecular Weight Determination of Glyoxalase II by Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis 57

    12. Inhibition of Glyoxalase I by GTP, ATP, CTP and UTP 60

    vii

  • F i

  • F i c? u r e Page

    25. Effect of Various Fixed Concentrations of Colchicine on the Glyoxalase I Activity at Increasing Levels of Free Glutathione with a Constant Level of Hemimercaptal . . 96

    26. Effect of Various Fixed Concentrations of Colchicine on the Glyoxalase I Activity at Increasing Levels of Free Methylglyoxal with a Constant Level of Hemimercaptal . . 99

    27. Inhibition of Glyoxalase II by the Hemimercaptal of Glutathione and Methylglyoxal 101

    IX

  • INTRODUCTION

    The conversion of phenylglyoxal to mandelic acid, when

    rabbits were fed phenylglyoxal, led to the discovery of the

    glyoxalase system in 1913 (1). Subsequently, the conversion

    of phenylglyoxal and methylglyoxal (pyruvaldehyde) to

    mandelic acid and lactic acid respectively was found in a

    variety of animal and plant tissues, as well as in yeast

    (2-4). At the time, the glyoxalase system was thought to be

    involved in glycolysis, but Lohman demonstrated that the

    glyoxalase system is not an essential part of glycolysis,

    and that the system requires glutathione as a cofactor (5).

    It was determined that glutathione, y-glutamyl-L-

    cysteinylglycine, can be replaced as a coenzyme by

    a-L~glutamyl-^-cysteinylglycine, B-^-aspartyl-L-cysteinyl-

    glycine or Y-D-glutamyl-L-cysteinylglycine, though all are

    less effective than glutathione (6,7). Other compounds

    tested as coenzymes but found ineffective include oxidized

    glutathione, cysteine, thioglycolic acid (5), Y-glutamyl-

    cysteine and cysteinylglycine (8). Wieland et al. concluded

    that glutathione derivatives which serve as the cofactor for

    the glyoxalase system must possess a terminal free amino

    group (9). This was based on the observations that

    N-acetylglutathione is not effective as a cofactor, but that

  • N-DL-alanylglutathione and N-DL-valylglutathione could serve,

    though less effectively than glutathione.

    Methylglyoxal can be replaced by other aliphatic and

    aromatic a-ketoaldehydes such as glyoxal, kethoxal, hydroxy—

    pyruvaldehyde, phenylglyoxal and some substituted phenyl-

    glyoxals (10,11). The ability of the a-ketoaldehydes to

    serve as substrates for the glyoxalase system is impaired

    when the side-group of the a-ketoaldehyde is sterically

    crowded. 2,4,6-Trimethylphenylglyoxal is not a substrate

    even though the related compound 2,4-dimethylphenylglyoxal

    does function as a substrate (11).

    In 1951 Racker showed that the glyoxalase system

    consists of two enzymes (12), glyoxalase I (EC 4.4.1.5) and

    glyoxalase II (EC 3.1.2.6). These two enzymes catalyze the

    following reactions with methylglyoxal as the substrate:

    0 O glyoxalase I

    CH3-C-CH + GSH

    (methylglyoxal) (glutathione)

    0 OH glyoxalase II

    CH3-C-CH-SG

    (S-lactoylglutathione) B^O

    OH I

    CH -CH-COOH + GSH

    (lactic acid)

  • It has been demonstrated that the glyoxalase system

    forms stereospecifically S-g-lactoylglutathione and D-lactic

    acid (13-16). The crucial step in the mechanism of glyox-

    alase I action was found to be the transfer of a hydride

    equivalent from C—1 to C—2 of the Oi—ketoaldehyde with the

    resulting thioester formation (17,18). Hal et al. intro-

    duced NMR evidence to support an enediol-proton transfer

    mechanism for this step (19).

    Davis and Williams showed that Mg is required by

    glyoxalase I from calf liver, but may not be necessary for

    the enzyme from yeast (20). Arronson et al. concluded using

    atomic absorption spectrometry, that glyoxalase I from human

    and porcine erythrocytes, as well as rat liver and yeast are

    zinc-metalloenzymes (21).

    Steady-state kinetics of glyoxalase I have been a con-

    troversial subject, and two independent studies have given

    rise to two different mechanisms for the action of

    glyoxalase I. Kermack and Matheson proposed a two substrate

    mechanism where free methylglyoxal and free glutathione are

    the two substrates (22). On the other hand, Cliffe and

    Waley supported a one substrate mechanism in which the true

    substrate is a hemimercaptal formed nonenzymatically from

    methylglyoxal and glutathione (23), as indicated below:

  • Methylglyoxal + glutathione k hemimercaptal

    glyoxalase I

    hemimercaptal > S-g-lactoylglutathione

    Subsequently, a third mechanism has been proposed which

    is a combination of the first two (24). This random-pathway

    mechanism involved one- and two-substrate branches. The

    two-substrate path involved glutathione and methylglyoxal as

    the first and second substrates, respectively. The

    one-substrate path utilized the hemimercaptal as the sole

    substrate. Except at very low hemimercaptal and free

    glutathione concentrations, the one—substrate pathway was

    proposed to predominate. At higher hemimercaptal and free

    glutathione concentrations the one-substrate pathway model

    predicts a hemimercaptal-free glutathione competition.

    Although the glyoxalase system has been known for many

    years and has received considerable attention from various

    investigators, the basic biological function of this system

    remains to be elucidated. The possibility that the

    glyoxalase system could be a step in carbohydrate metabolism

    was eliminated by Lohman (5) by showing that the system is

    not an essential step in glycolysis. It has been suggested

    that the glyoxalase system may function metabolically by

    providing a mechanism for the entry of hydroxypyruvic

    aldehyde into the tricarboxylic acid cycle or the glycolytic

  • pathway (25). Support for this was the finding that

    phosphohydroxypyruvic aldehyde is a substrate for glyoxalase

    I, and the corresponding glutathione thioester could be

    hydrolyzed by glyoxalase II to form D-3-phosphoglyceric

    acid. However, there is no evidence for the natural occur-

    rence of phosphohydroxypyruvic aldehyde (26).

    It has also been postulated that the glyoxalase system

    may participate in a cycle for the degradation of glycine

    and threonine (27-29) via aminoacetone in view of the fact

    that methylglyoxal is the deamination product of aminoace-

    tone. Enzymatic conversion of aminoacetone to methylglyoxal

    was first found in ox plasma, but subsequent studies found

    this conversion in only a few other biological systems (30).

    Glutathione and the glyoxalase system may possibly play

    some role in the regulation of porphyrin synthesis by con-

    version of some part of Y,6—dioxovalerate into tricarboxylic

    acid cycle intermediates, y,6-Dioxovalerate has been found

    to be a substrate for glyoxalase I and the resulting gluta-

    thione-thioester is then hydrolyzed by glyoxalase II to form

    D-a-hydroxyglutarate (31).

    It has also been postulated that a-ketoaldehydes might

    have a special function as regulatory molecules in cell

    division (32-35). Szent-Gyorgyi et al. defined "retine" as

    the agent that prevents cell division and it was proposed to

    be a derivative of methylglyoxal. "Promine" was defined as

  • the agent which controls the action of "retine", and the

    glyoxalase system can serve as "promine". This view, along

    with the known cancerostatic and cytotoxic properties of

    methylglyoxal (36-39), gave rise to the hypothesis that

    selective inhibition of glyoxalase I should provide a means

    of retarding cell division. This approach has been taken by

    Vince and his co-workers (40-42), who have synthesized a

    large number of S-substituted glutathione derivatives as

    potential inhibitors of glyoxalase I and tested them with

    the yeast enzyme. The most effective compounds were those

    containing long S-alkyl chains or nonpolar S-aryl groups.

    Methylglyoxal was once believed to be involved in

    glucose catabolism as a component of the so-called non-

    phosphorylating glycolysis (43). When phosphorylated

    compounds were identified as the intermediates in glucose

    breakdown, the formation of methylglyoxal was considered to

    be due to nonenzymatic side-reactions and thus of little or

    no importance (44). Methylglyoxal gained more importance

    after the reports on an enzyme from Escherichia coli,

    Pseudomonas saccharophila and Proteus vulgaris which

    catalyzes the conversion of dihydroxyacetone phosphate to

    methylglyoxal and inorganic phosphate (15,45,46). This

    enzyme, methylglyoxal synthase, appears to constitute the

    first step of a glycolytic bypass from triosephosphates to

    pyruvate via methylglyoxal and g-lactate. Methylglyoxal

  • synthase is not detected in liver, skeletal muscle, and

    brain of the rat, nor in erythrocytes, lymphocytes, skeletal

    muscle, kidney, liver, brain and cardiac muscle of the

    human, or in other species, including crab, crayfish,

    shrimp, ascarid, snail, lizard, frog, clam, starfish,

    jellyfish, sponge, yeast (Saccharomyces cerevisiae and

    Candida albicans), algae (Chlorella sp.), fungi (Aspergillus

    sp., Trichophytan sp. and Penicillum sp.) and Euglena

    gracilis (47). The absence of methylglyoxal synthase in

    these organisms and tissues raises a question as to the role

    of the glyoxalase system, since it is present in the tissues

    or organisms apparently lacking methylglyoxal synthase.

    Two recent reports provide evidence that methylglyoxal

    is most probably the true substrate for the glyoxalase

    system (48,49). Methylglyoxal was isolated from beef liver

    as its 2,4-dinitrophenylhydrazone and identified by com-

    parison to synthetic methylglyoxal (48). Sato et al. (49)

    concluded that methylglyoxal formation in rat liver cells is

    a metabolic process, and is largely derived from dihydroxy-

    acetone phosphate.

    Over the years, glyoxalase I has received considerable

    attention from numerous investigators. The first attempt to

    purify glyoxalase I was by ammonium sulfate precipitation of

    crude tissue homogenates (2). Glyoxalase I has been

    partially purified from calf liver (50), from livers of

  • 8

    normal DBA/lJ mice and from the same mice bearing a lympho-

    sarcoma (51), from the livers of rats of various ages (52),

    and from porcine erythrocytes (53). Nearly homogeneous

    preparations of glyoxalase I have been obtained from rat

    liver, erythrocytes, brain and kidney (11); homogeneous

    preparations from mouse liver (54,55) and from rat liver

    (56) have been reported in the literature.

    A nearly homogeneous preparation of glyoxalase II from

    human liver with a very low yield has been reported by

    Uotila (57). Recently, glyoxalase II has been purified to

    homogeneity from mouse liver and rat erythrocytes (58,59).

    Some recent reports in the literature have suggested

    that the glyoxalase enzymes are involved in the polymer-

    ization of tubulin to form microtubules (60,61,62). The

    microtubules are proteinaceous organelles that are present

    in virtually all eucaryotic cells. The microtubules are

    composed of subunits assembled into elongated tubular

    structures having an indefinite length? they are capable of

    rapid changes in length by assembly or disassembly of the

    subunit proteins, the tubulins, by such conditions as tem-

    perature changes, hydrostatic pressure changes, and by

    certain drugs such as colchicine and vinblastine. The

    microtubules are associated along with other proteins with

    the formation of complex assemblies such as the mitotic

    spindle, centrioles, cilia and flagella, axonemes, and

  • neurotubules, and are a determinant of cell shape and

    motility (63).

    Colchicine, the best known of all microtubule poisons is

    believed to combine at a specific site with the tubulin

    dimer (M.W. 110,000) in a 1:1 ratio (64). It has been found

    that colchicine binds to tubulin and prevents the in vitro

    assembly of microtubules (65); whereas lumicolchicine, the

    irradiation product of colchicine, neither binds to tubulin

    nor prevents microtubule assembly. Vinblastine, one of the

    Vinca alkaloids, which in very low concentrations causes

    precipitation of tubulin granules (thereby inhibiting

    assembly), has a binding site on tubulin which is different

    from that of colchicine (64). Additionally, the two binding

    sites on tubulin for the guanosine nucleotides are distinct

    (64). The colchicine binding site on tubulin is also the

    binding site for the microtubule poison, podophyllotoxin

    (66). Another drug, griseofulvin, does not apparently block

    the in vitro polymerization of tubulin as measured by

    electron microscopy (66); this drug may not therefore

    directly affect microtubules, but may act on other processes

    essential for normal completion of cell division.

    There were several objectives of the studies reported

    herein. The first objective was to devise a simple,

    convenient, and reproducible method of methylglyoxal prepa-

    ration, since methylglyoxal is very unstable even stored

  • 10

    under nitrogen in the cold. Consequently, the tedious and

    cumbersome preparation and standardization of methylglyoxal

    solutions have been periodically required when continuing

    glyoxalase I assays were being conducted.

    A second objective was to develop new schemes for the

    purifications of both glyoxalase enzymes employing affinity

    chromatography as a major step. In this manner, it was

    hoped that higher overall yields could be obtained, and any

    alteration of enzyme protein resulting from the harsh

    methods previously employed could be avoided.

    A third objective involved a comparative study of the

    structure and composition of the purified glyoxalase

    enzymes, employing chemical, physical and kinetic charac-

    terizations. It was anticipated that questions relating to

    a possible structural relationship between the two

    glyoxalases might be answered by the application of these

    methods.

    Finally, experiments were to be conducted which would

    bring additional insights as to the biological function of

    the glyoxalase system. One approach to be taken involved a

    search for cellular metabolites which might affect the

    activity of glyoxalases, either activators or inhibitors.

  • EXPERIMENTAL PROCEDURE

    General. Swiss mice (24-27 g) were purchased from

    Timco, Houston, Texas. Yeast glyoxalase I, hexokinase, glu-

    coses-phosphate dehydrogenase, glucose phosphate isomerase,

    a-glycerophosphate dehydrogenase, triosephosphate isomerase,

    aldolase, phosphofructokinase, g-lactate dehydrogenase,

    glutathione and oxidized glutathione were obtained from

    Sigma Chemical Co., St. Louis, MO. 3-Hydroxy-3-methyl-

    glutaryl-CoA reductase assays were conducted by Dr. R. E.

    Thompson and co-workers, glycogen synthase and glycogen

    phosphorylase assays by Dr. B. G. Harris and co-workers, and

    S-adenosylmethionine synthetase and cyclopropane fatty acid

    synthase assays by Dr. D. D. Smith. Pyruvaldehyde dimethyl

    acetal and alkylamines were purchased from Aldrich Chemical

    Co., Milwaukee, Wis.? semicarbazide•HC1 and imidazole were

    obtained from Eastman Organic Chemicals, Rochester, N.Y.

    Nucleotides, nucleosides and structurally related compounds

    were purchased either from Sigma Chemical Co., St. Louis,

    MO. or from Calbiochem, Los Angeles, CA.

    S-g-Lactoylglutathione was prepared and purified by the

    procedure of Uotila (67). S-Octylglutathione was prepared

    by reacting 1-bromooctane with glutathione employing Method

    A of Vince ejt a_l. (41). Lumicolchicine was prepared by

    11

  • 12

    irradiation of colchicine with ultraviolet light utilizing

    the method of Wilson and Friedkin (68).

    All other chemicals were the highest purity obtainable

    from commercial sources. All enzymes were assayed with

    the usual assay procedures obtainable from the literature.

    All chromatography columns were run at 4°, and enzyme

    preparations were stored at -30°.

    Methylglyoxal synthesis. The synthesis of methyl-

    glyoxal (pyruvaldehyde) was accomplished by the hydrolysis

    of pyruvaldehyde dimethyl acetal, as indicated below:

    0 H + 0 0 II II II

    CH.-C-CH(OCH0)0 > CH -C-CH

    Pyruvaldehyde dimethyl acetal (10.0-605 pi, 0.083-5.02

    mmol), 2.5 ml of 10% H2S°4' an

  • 13

    Alexander and Boyer (70) were employed to quantify

    methylglyoxal.

    Routine glyoxalase I assay. Glyoxalase I activity was

    determined by a modification of the procedure of Racker

    (12). The reaction mixture used was: 7.9 mM methylglyoxal,

    1.0 mM glutathione, 200 mM imidazole•HC1 (pH 7.0), and 16 mM

    MgSO^. The reaction mixture was allowed to stand for at

    least 10 minutes at room temperature to ensure equilibration.

    The hemimercaptal concentration was calculated to be 0.7 mM

    using K = 3.1 mM (41). The enzymatic production of

    S-g-lactoylglutathione ( E ^ = 3.37 mM - 1™" 1) was

    followed at 240 nm for 2 minutes at 25° on a Beckman DBG

    recording spectrophotometer. The reaction was initiated by

    the addition of the enzyme preparation (1-20 jal) to 3.0 ml

    of reaction mixture, and initial rates were determined by

    the slope of the linear portion of the plot. The reference

    cell contained all reaction mixture components with the

    exception of the enzyme preparation. A unit (I.U.) of

    glyoxalase I activity is defined as the amount of enzyme

    catalyzing the formation of 1 jumol of S-g-lactoylglutathione

    per minute in the routine enzyme assay system. Specific

    activity is expressed as units per mg protein.

    Routine glyoxalase II assay. The reaction mixture used

    was: 0.5 mM S-D-lactoylglutathione in 100 mM potassium

    phosphate (pK 7.0). The enzymatic disappearance of

  • 14

    S-D-lactoylglutathione ^240 ~ ^*37 ^ c m w a s

    followed at 240 nm for 5 minutes at 25° on a Beckman DBG

    recording spectrophotometer. The reaction was initiated by

    the addition of the enzyme preparation (5-75 jal) to 3.0 ml

    of reaction mixture, and the initial rates were determined

    by the slope of the linear portion of the plot. A unit

    (I.U.) of glyoxalase II is defined as the amount of enzyme

    catalyzing the conversion of 1 jumol S-D-lactoylglutathione

    per minute in the routine enzyme assay system.

    Determination of protein concentration. Protein con-

    centrations were determined by the colorimetric Coomassie

    Blue procedure of Bradford (71). Crystalline bovine serum

    albumin was used as the protein standard. The protein

    levels in the effluent of the chromatography columns were

    monitored by their absorbance at 280 nm.

    Column material preparation. The preparations of

    hydrophobic and affinity chromatography column materials

    were based on the method of Cuatrecasas (72). To 100 ml of

    washed Sepharose 4B, 25 g of finely divided cyanogen bromide

    was added with stirring. The pH was maintained at 10.5 by

    the addition of 5 N NaOH. The temperature was maintained

    between 18 to 20° by the addition of ice to the reaction

    mixture. When all cyanogen bromide was dissolved (12-15

    minutes), the activated Sepharose was quickly washed with a

    minimum of 10 volumes of cold 0.1 M sodium carbonate (pH

  • 15

    10.2). The step of washing the activated Sepharose was

    accomplished in less than 90 seconds. For hydrophobic

    chromatography columns, the appropriate n-alkylamine (3.6

    mmoles) which was dissolved in 100 ml of wash buffer was

    then added with gentle stirring to the activated Sepharose.

    The mixture was stirred overnight at 5°. It was then

    washed extensively with distilled water and placed in the

    buffer used for chromatography. For the preparations of

    affinity chromatographic column materials, oxidized

    glutathione and S-octylglutathione were coupled to Sepharose

    under the same conditions. The degree of substitution of

    the ligands was not determined.

    Purification of glyoxalase I. Swiss mice were

    sacrificed either by asphyxiation in CO^ or by cervical

    dislocation. The livers were immediately removed and

    homogenized (0 for 45-60 seconds at medium speed with a

    Virtis homogenizer) in three volumes of a solution of 1 mM

    potassium phosphate (pH 7.0), 1 mM MgS04 and 20% glycerol.

    The homogenate was centrifuged at 100,000 x g for 1 hour.

    The supernatant fraction thus obtained (approximately 225 ml

    per 100 g liver) was designated as the crude preparation of

    glyoxalase I and contained 25-30 mg protein/ml. When the

    livers were not used immediately they were frozen in dry ice

    and then stored at -30°.

  • 16

    At the outset of glyoxalase I purification, sufficient

    crude preparation to give 6.2 grams of protein was placed on

    a 2.5 x 45 cm ethylamine-Sepharose (hydrophobic) column. The

    column material had been previously equilibrated with a

    solution of 1 mM potassium phosphate (pH 7.0), ImM MqSO 4

    and 20-6 glycerol. After loading, the column was washed with

    the equilibration solution until the concentration of

    protein being eluted was quite low (monitored at 280 nm). A

    linear phosphate gradient (1-50 mM, pH 7.0, a total of 7

    liters) in 5 mM MgSO^ and 20% glycerol was used to elute

    glyoxalase I. The phosphate concentrations were determined

    by the method of Chen et al. (73).

    Pooled active fractions from the ethylamine-Sepharose

    column were then added to a 1.5 x 20 cm S-octylglutathione-

    Sepharose affinity chromatographic column which had been

    equilibrated with a solution of 10 mM potassium phosphate

    (pH 7.0), 5 mM MgSC>4 and 20% glycerol. After sample

    application, the column was washed with the equilibration

    solution until no protein could be detected in the effluent.

    The column was then treated with 400 ml of 50 mM imidaz-

    ole *HC1 (pH 7.2) solution containing 5 mM MgSO^ and 20%

    glycerol. Homogeneous glyoxalase I was then eluted by the

    addition of the above imidazole buffer medium containing 20

    mM glutathione.

  • 17

    Purification of glyoxalase II. Swiss mice were sacri-

    ficed by cervical dislocation. The livers were immediately

    removed and homogenized (0° for 2 minutes at medium speed

    with a Virtis homogenizer) in two volumes of a solution of

    10 mM potassium phosphate (pH 7.0) and 20% glycerol. The

    homogenate was centrifuged at 100,000 x g for 1 hour. The

    supernatant fraction thus obtained was designated as the

    crude preparation of glyoxalase II. When the livers were

    not used immediately they were frozen in dry ice and stored

    at -30°.

    Sufficient crude preparation to give 7.0 grams of

    protein was placed on a 2.6 x 70 cm oxidized glutathione-

    Sepharose affinity chromatographic column. The column

    material had been previously equilibrated with a solution of

    10 mM potassium phosphate (pH 7.0) and 20% glycerol. After

    loading, the column was washed with the equilibration

    solution until the bulk of the protein was eluted. The

    column was then washed with 350 ml of a solution of 50 mM

    potassium phosphate (pH 7.0) and 20% glycerol, and again

    with the equilibration solution (600 ml). A gradient of a

    competitive inhibitor of glyoxalase II, S-octylglutathione,

    (1 mM to 5 mM, a total of 2 liters) prepared in the equil-

    ibration solution, was used to elute glyoxalase II.

    Pooled active fractions from the first column were

    diluted two-fold with the equilibration solution. This

  • 18

    diluted enzyme preparation was then added to a 2.6 x 40 cm

    oxidized glutathione-Sepharose affinity chromatographic col-

    umn, which had been previously equilibrated with a solution

    of 10 mM potassium phosphate (pH 7.0) and 20% glycerol.

    After sample application the column was washed with the

    equilibration solution/ then with 150 ml of a solution of 75

    mM potassium phosphate (pH 7.0) containing 20% glycerol, and

    again with the equilibration solution (250 ml). Homogeneous

    glyoxalase II was then eluted by addition of a solution of

    2.0 mM S-octylglutathione in the equilibration solution.

    Concentration procedures. The pooled active fractions

    from the affinity columns employed for the purifications of

    both enzymes were placed in a Millipore 142 mm Hi-Flux Cell

    ultrafiltrator. The enzymes were concentrated to a volume

    of approximately 50 ml. Further concentrations of the

    enzyme preparations were carried out with Millipore

    immersible CX ultrafiltration units which have a molecular

    weight cut-off of 10,000 daltons.

    Molecular weight determination of glyoxalase I by

    sedimentation equilibrium. Sedimentation equilibrium exper-

    iments were conducted in a Beckman-Spinco Model E analytical

    ultracentrifuge equipped with RTIC temperature control and

    electronic speed control. The meniscus depletion method

    (74,75) was used in the An-D rotor in a 12 mm double sector

    cell with sapphire windows. Glyoxalase I (0.420 mg/ml) was

  • 19

    extensively dialyzed in K H2 P°4' 9/1* Na^HPO^, 4.3

    g/l (pH 7.413); P=1.0020. Centrifugation was carried out

    for 30 hours at speeds of 28,000 and 32,000 rpm.

    Molecular weight determinations of glyoxalase enzymes

    by sodium dodecyl sulfate polyacrylamide gel electro-

    phoresis. For the molecular weight determinations of

    glyoxalase I the method reported by Fairbanks et al. (76)

    was employed. Gels were prepared with a 5.6% polyacrylamide

    concentration in a solution of 40 mM Tris-HCl (pH 7.4), 20

    mM sodium acetate, 2 mM EDTA and 1% sodium dodecyl sulfate,

    and stained with 0.2% Coomassie Blue-R in an acetic acid/

    methanol/water (8:46:46, v/v) solution. After staining for

    at least 2 hours, the gels were destained in an acetic

    acid/methanol/water (10:45:45, v/v) solution. Bovine serum

    albumin (67,000), ovalbumin (45,000) and ribonuclease A

    (13,700) were used as standards for molecular weight deter-

    minations . Standards and samples were dialyzed against a

    solution composed of 10 mM Tris-HCl (pH 8.0), 1 mM EDTA and

    0.1% 2-mercaptoethanol, they were made 2% in sodium dodecyl

    sulfate and incubated at 100° for two minutes, before the

    electrophoresis run.

    For the molecular weight determinations of glyoxalase

    II the method of Maizel (77) was employed. Gels were

    prepared with a 7.5% polyacrylamide concentration in a

    solution of 200 mM sodium phosphate (pH 7.0) and 1% sodium

  • 20

    dodecyl sulfate, and stained with 0.2% Coomassie Blue-R in

    an acetic acid/methanol/water (8:46:46, v/v) solution.

    After staining for at least 2 hours, the gels were destained

    in an acetic acid / methanol / water (10:45:45, v/v)

    solution. Ovalbumin (45,000), glyceraldehyde-3-phosphate

    dehydrogenase (37,000), triose phosphate isomerase (26,500)

    and hemoglobin (16,500) were used as standards for molecular

    weight determinations. Before the electrophoresis runs,

    samples and standards were dialyzed against 200 mM sodium

    phosphate (pH 7.0) and 0.1% 2-mercaptoethanol, they were

    made 2% in sodium dodecyl sulfate and incubated at 100°

    for two minutes.

    Polyacrylamide gel electrophoresis. Polyacrylamide gel

    electrophoresis was performed according to the method of

    Maizel (77). Gels were prepared with a 7.5% polyacrylamide

    concentration in a solution of 2.5 mM Tris • HC1, 20 mM

    glycine (pH 9.0). The staining and destaining of the gels

    were carried out by the same procedures as described above

    for the sodium dodecyl sulfate polyacrylamide gel electro-

    phoresis experiments.

    Isoelectric focusing. Isoelectric focusing was per-

    formed in a LKB Multiphor Electrophoresis Unit according to

    the instructions of the manufacturer. A pH gradient of

    3.5-10 was used with 1% (w/v) ampholyte concentration.

  • 21

    Kinetic studies. It was assumed that the hemimercaptal

    of methylglyoxal and glutathione is the true substrate of

    glyoxalase I, and that the dissociation constant for the

    hemimercaptal is 3.1 mM (41). The appropriate amounts of

    both methylglyoxal and glutathione are added together to

    form, nonenzymatically, the required hemimercaptal

    concentration before the addition of the enzyme. Since

    equilibrium solutions of hemimercaptal would also contain

    glutathione and methylglyoxal, the concentrations of

    glutathione and methylglyoxal after equilibration are

    designated as free glutathione and free methylglyoxal.

    Therefore, experiments utilizing hemimercaptal will state

    not only hemimercaptal concentration, but also either free

    glutathione or free methylglyoxal levels. For the exper-

    iments involving the inhibition of glyoxalase II by the

    hemimercaptal, the free glutathione level was 0.3 mM. This

    level of glutathione does not inhibit the human glyoxalase

    II (57).

  • RESULTS AND DISCUSSION

    Methylglyoxal Synthesis. Methylglyoxal, for use in en-

    zymatic studies, is usually obtained by vacuum (54) or steam

    distillation (10) of a commercial 40% solution in order to

    eliminate polymerized methylglyoxal. The pale green distil-

    late may then be passed through an anion exchange resin

    (54,78) to obtain acid-free methylglyoxal. Methylglyoxal is

    then standardized by Friedemann's titration method (69)

    after oxidation with H2°2' o r enzYmatically (79), or by

    the semicarbazide method of Alexander and Boyer (70). This

    tedious method of distillation, removal of acidic contami-

    nants, and quantitation is time consuming. Furthermore,

    methylglyoxal is very unstable and readily undergoes

    polymerization and oxidation reactions (80). The true

    methylglyoxal content of solutions stored for even short

    time periods is questionable at best.

    For studies involving repetitive assays of glyoxalase

    I, a simple, convenient, and reproducible method for the

    preparation of methylglyoxal is needed. The rapid and

    direct method for the conversion of pyruvaldehyde dimethyl

    acetal to methylglyoxal (81), which does not require a

    quantitation analysis with each preparation, serves as a

    22

  • 23

    convenient procedure for the preparation of methylglyoxal

    for use in glyoxalase I assays.

    The effect of temperature and time on the hydrolysis of

    the acetal are shown in figures 1A and IB respectively.

    From figure 1A, it is apparent that the hydrolysis proceeds

    at an appreciable rate only at temperatures approaching

    100°. As can be seen (Figure IB), the concentration of the

    methylglyoxal produced (disemicarbazone assay) is highest

    after a 25 minute hydrolysis. This time period was used in

    all subsequent hydrolysis procedures. As subsequently

    indicated, the hydrolysis reaction is virtually quantitative

    when the process is carried out at 100° for 25 minutes.

    The effect of H 2S°4 c o n c e n t r a tion on the hydrolysis of

    pyruvaldehyde dimethyl acetal is shown in Figure 2. Five

    percent H 2S 0

    4 w a s chosen as the acid concentration for the

    hydrolysis, since this was the lowest concentration that

    gave essentially quantitative hydrolysis under the experi-

    mental conditions employed.

    The effect of increasing pyruvaldehyde dimethyl acetal

    concentrations on the extent of hydrolysis was studied. As

    shown in Figure 3, the response plot is linear up to an

    acetal concentration of at least 450 mM. Each point in the

    plot represents essentially a quantitative conversion to

    methylglyoxal. It is probable that even higher levels of

  • 24

    Figure 1

    A. Effect of Temperature on Hydrolysis of Pyruvaldehyde

    Dimethyl Acetal

    Pyruvaldehyde dimethyl acetal (200 pi, 1.65 mmole) was

    hydrolyzed in a final volume of 5.0 ml of 5% H SO for 2 4

    30 minutes. Ten pi of a 1:50 dilution of the hydrolysate

    was added to 3.0 ml of 0.067 M semicarbazide•HC1 in 0.1 M

    sodium phosphate ( PH 7.4). After 15 minutes, the absorbancy

    at 286 nm was used to determine the methylglyoxal concen-

    tration.

    B. Rate of Hydrolysis of Pyruvaldehyde Dimethyl Acetal

    Pyruvaldehyde dimethyl acetal (200 pi, 1.65 mmole) was

    hydrolyzed in a final volume of 5.0 ml of 5% H SO at ° _ 2 4

    00 c. The conditions used to determine methylglyoxal

    concentrations are given in the legend of figure 1A.

  • 25

    t CD

    i in *

    a 00

    i CM •

    1 T~ •

    o • o o* o o O 9 9 2 w

    Z

    2

    o o

  • 26

    Figure 2

    Effect of Acid Concentration on Hydrolysis of

    Pyruvaldehyde Dimethyl Acetal

    Pyruvaldehyde dimethyl acetal (150 pi, 1.24 mmole) was

    hydrolyzed in a final volume of 5.0 ml at 100°C for 25

    minutes in the presence of varying levels of . '^ie

    conditions used to determine methylglyoxal concentrations

    are given in the legend of figure 1A.

  • 27

    286

    6 * H2SO4

  • 28

    Figure 3

    Hydrolysis of Varying Levels of Pyruvaldehyde

    Dimethyl Acetal

    Each point represents a separate hydrolysis process. The

    abscissa represents the acetal concentration in the hydro-

    lysis mixture. The ordinate represents the methylglyoxal

    concentration formed after the hydrolysis. Hydrolysis

    experiments were carried out at 100°C for 25 minutes in a

    final volume of 5.0 ml of 5% H2SC>4. The conditions used

    to determine methylglyoxal concentrations are explained in

    the legend of figure 1A.

  • 29

    £

    %

    o LU

    X cr o LL

    O

    100 200

    [ACETAL] , mM

  • 30

    acetal can be employed with a similar stoichiometric

    conversion resulting.

    In a separate study, triplicate samples from each of

    six individual hydrolysates were quantitated for methyl-

    glyoxal concentration by the semicarbazide method. The

    original concentration of pyruvaldehyde dimethyl acetal in

    each hydrolysis flask was 330 mM. After hydrolysis, the

    concentration of the resulting methylglyoxal was found to be

    329 mM (S.D. = +7). From these data it is apparent that

    hydrolysis goes to completion.

    Methylglyoxal prepared by this procedure was used for

    the glyoxalase I kinetic studies. Two separate hydrolyses

    and enzyme assays were conducted on two consecutive days,

    and the response plot shown in Figure 4 is a composite of

    those two experiments. It is obvious that a single curve

    fits the data points obtained from both experiments.

    In kinetic studies using glyoxalase I, substrate con-

    centrations are usually calculated from the dissociation

    constant of the hemimercaptal (41). The required substrate

    concentrations are prepared by the addition of appropriate

    amounts of both methylglyoxal and glutathione. It has been

    found that methylglyoxal is very unstable, even when stored

    under nitrogen in the cold. Consequently, the tedious and

    cumbersome preparation, removal of acidic contaminants and

    standardization of methylglyoxal solutions have been

  • 31

    Figure 4

    Glyoxalase I Kinetic Study with Methylglyoxal Prepared

    by the Hydrolysis of Pyruvaldehyde Dimethyl Acetal

    The abscissa values represent the hemimercaptal concen-

    tration derived from the equilibrium mixture of glutathione

    and methylglyoxal. For each data point represented, the

    methylglyoxal was obtained from separate hydrolyses of

    pyruvaldehyde dimethyl acetal (varying concentrations).

    Hydrolysate (0.25 ml) and an appropriate amount of glu-

    tathione were diluted to a final volume of 10.0 ml. The

    free glutathione level after the nonenzymatic equilibrium

    reaction was 0.3 mM (see Experimental Procedure). Homo-

    geneous glyoxalase I (4 I.U.) was used as enzyme source in

    the assays. The plot is a composite of two separate

    experiments conducted on consecutive days. • represents the

    data obtained the first day; #the second day.

  • 32

    a 0 I# k 9 E 1 o z

    (U|IU/VV)A

  • 33

    periodically required when continuing glyoxalase I assays

    are being conducted. This method of stoichiometric

    hydrolysis of pyruvaldehyde dimethyl acetal to methylglyoxal

    is simple, reproducible, and permits the quick preparation

    of fresh solutions of methylglyoxal of known concentrations

    on a daily basis.

    Purification of glyoxalase I. The purification of

    Swiss mouse liver glyoxalase I was accomplished by use of

    both hydrophobic and affinity chromatography. The choice of

    a specific hydrophobic grouping for the first purification

    step was made after a study of a variety of alkyl chains

    covalently linked to Sepharose 4B. As the length of the

    hydrophobic chain is increased, glyoxalase I binds more

    tenaciously, and the concentrations of phosphate or imidaz-

    ole buffers necessary to elute the enzyme increases greatly

    (Table I). With hydrophobic columns prepared with alkyl

    chains of six carbons or less, hemoglobin and other unde-

    sired proteins were not bound, but passed through in the

    washings. The ethylamine-containing hydrophobic column was

    therefore chosen for use in the purification of glyoxalase

    I, since the enzyme could be eluted at low ionic strengths

    and much of the contaminating protein was not initially

    bound, giving better purifications. The glyoxalase II

    activity was eluted with the early fractions containing the

    bulk of the protein.

  • 34

    TABLE I

    HYDROPHOBIC CHROMATOGRAPHY OF GLYOXALASE I

    b Eluting Agent

    Liganda Liganda

    Imidazole(M) Phosphate(M)

    Ethylamine 0.02

    n-Propylamine 0.1 0.08

    n-Butylamine 0.4 0.2

    n-Pentylamine 0.7 0.8

    n-Hexylamine 1.0 1.5

    n-Octylamine 2.0 -

    n-Decylamine 4.0 —

    Details of coupling of alkylamines to CNBr-activated

    Sepharose are given in Experimental Procedure.

    b The concentrations of imidazole and phosphate buffers

    necessary to elute glyoxalase I from the corresponding

    column materials.

  • 35

    A variety of glyoxalase I inhibitors which could

    possibly serve as affinity chromatography ligands have been

    prepared (20,41,82-84). S-(10-Aminodecyl)glutathione has

    been successfully employed as a ligand for the affinity

    chromatographic purification of glyoxalase I from DBA/lJ

    mouse liver (55). Unfortunately, affinity columns prepared

    with this ligand have low capacities for glyoxalase I. Fur-

    thermore, these columns cannot be used repeatedly on crude

    preparations because of the degradation of the glutathione

    moiety by endogeneous glutathionase activity. In an effort

    to minimize the latter problem, glutaryl-S-(10-aminodecyl)-

    ^-cysteinylglycine (83) was studied as a possible ligand for

    affinity chromatography, but it was found to be totally

    ineffective. It was therefore concluded that the covalent

    linkage of the ligand to Sepharose should be with an -amino

    group (not present in the glutaryl-containing compounds) for

    effective affinity chromatography. S-Octylglutathione, a

    potent inhibitor of glyoxalase I (41), was studied as an

    affinity ligand and was found to be quite effective. The

    capacity of the column material to bind glyoxalase I

    activity is high, and the enzyme is specifically eluted in

    good yield by additions of glutathione. A single pass of

    the active fractions from the hydrophobic chromatography

    through the S-octylglutathione-Sepharose column was

    sufficient to purify glyoxalase I to homogeneity.

  • 36

    Table II summarizes the purification data of Swiss

    mouse liver glyoxalase I. The 1162-fold purified enzyme

    exhibited a specific activity of 944 units per mg protein.

    The enzymatic activity has been shown to be associated with

    a single protein species which has been purified to apparent

    homogeneity. The above purification factor is based on a

    100,000 x g supernatant fraction obtained from a crude cell

    homogenate.

    Figure 5 shows the protein concentration, the

    glyoxalase I activity and the phosphate concentration

    profiles obtained on an ethylamine-Sepharose hydrophobic

    chromatographic column. The phosphate concentrations were

    determined by the method of Chen et al̂ . (73). A phosphate

    gradient was used to elute glyoxalase I activity. It should

    be noted that better purifications were achieved by gradient

    elution than by increasing the ionic strength in a stepwise

    manner.

    Affinity chromatography using S-octylglutathione as the

    immobilized ligand proved to be invaluable as a purification

    step. Figure 6 shows the protein concentration and

    glyoxalase I activity profiles obtained on an S-octyl-

    glutathione-Sepharose affinity chromatographic column. To

    elute the glyoxalase I activity from the affinity column, 20

    mM glutathione was required. This concentration of an

    eluant may be considered quite high; however,

  • w h-3 CQ <

    H w CO < 1

    3 s o

    fa o

    £ o H Eh C u H fa H PS P fa

    u 0 •p rH 0 rH 0 0 •H -P .p •H 03

    o >i cn

    ftUH

    >1 -P

    37

    CN • O CN

    V0

    CN KO

    o CN VQ • • •

    1—1 o CN LO

    CN cr>

    i—i 00 v£> • • •

    o V0 CO 1—1 as

    •H • CO ĉ rH > D 00 00 r-*H • 00 r̂ ID •P H CN CN 0 —' <

    d) E — o o LO 3 H CN 00 H £ CM CN in 0 — rH >

    3 a) c rH c •H Cn

    0 Q* e r-H *H (d >1 -P 0) rH •P 0 >1 0 fd rC O

    -P 1 fa u W CO

  • 38

    Figure 5

    Ethylamine-Sepharose Hydrophobic Chromatography of

    Swiss Mouse Liver Glyoxalase I

    Experimental details of chromatography and enzyme assays are

    described in Experimental Procedure. Crude preparation (220

    ml) was applied to a 2.5 x 45 cm preequilibrated ethylamine-

    Sepharose column. Fraction volumes were 20 ml. The arrow

    marks the application of potassium phosphate gradient (1-50

    mM, pH 7.0). o — o , glyoxalase I activity, AA/min (20 pi of

    each fraction was used in the assay); • •, protein concen-

    tration, mg/ml; x — x , phosphate concentration, mM.

  • 39

    P R O T E I N ( m g / m l )

    ( u | u i / w )

    A l l A i l D V I 3 S V 1 V X O A 1 9

  • 40

    Figure 6

    S-Octylglutathione-Sepharose Affinity Chromatography

    of Swiss Mouse Liver Glyoxalase I

    Experimental details of chromatography and enzyme assays are

    described in Experimental Procedure. Pooled fractions (1280

    ml) from the hydrophobic chromatography column were applied

    to a 1.5 x 20 cm preequilibrated S-octylglutathione-

    Sepharose column. Fraction volumes were 6.9 ml. The arrow

    marks the application of glutathione (20 mM). o — o , glyox-

    alase I activity, AA/min (10 pi of each fraction was used in

    the assay); • — • , protein concentration, jag/ml.

  • 41

    PROTEIN ( p g / m l ) O 0 O

    N

    * CJ O O

    (UJU3/VV) A1IAI1DV I 3SV1VX0A10

  • 42

    S-octylglutathione is a very potent inhibitor of glyoxalase

    I (41), and it would be expected that relatively high

    concentrations of glutathione would be needed to elute the

    enzyme.

    Polyacrylamide gel electrophoresis and sodium dodecyl

    sulfate polyacrylamide gel electrophoresis were conducted on

    the purified glyoxalase I. The amount of the enzyme applied

    to the gels varied from 8 to 20 jig protein. The gels ex-

    hibited only one band with both electrophoretic procedures;

    no minor bands could be detected either visually or by

    densitometric tracings of the gels.

    In other studies, attempts were made to use affinity

    chromatography as a one-step purification procedure starting

    with the crude preparation; purifications up to 240 fold

    have been achieved. Modifications of absorption and elution

    conditions could be further investigated to improve this

    purification factor.

    Purification of glyoxalase II. The purification of

    Swiss mouse liver glyoxalase II was accomplished by use of

    affinity chromatography. Some glyoxalase I inhibitors, such

    as various S-alkyl glutathiones, were studied as possible

    affinity ligands. The column materials prepared with these

    compounds do bind the glyoxalase II activity, but it has

    been found that effective elution of the enzyme was not

    possible. Oxidized glutathione was studied as an affinity

  • 43

    ligand and was found to be quite effective. Affinity

    columns with this ligand readily bound glyoxalase II and had

    a high capacity. The enzyme was selectively eluted with the

    addition of 2 mM S-octylglutathione, a linear competitive

    inhibitor of glyoxalase II (K^ =1.5 mM). The passing of

    the enzyme preparation through the same kind of affinity

    chromatographic column two times was sufficient to purify

    glyoxalase II to homogeneity.

    Table III summarizes the purification data of Swiss

    mouse liver glyoxalase II. The overall yield obtained was

    80% with a 2479-fold purification, and the enzyme exhibited

    a specific activity of 920 units per mg protein. The

    enzymatic activity has been shown to be associated with a

    single protein species which has been purified to apparent

    homogeneity. The above purification factor is based on a

    100,000 x g supernatant fraction obtained from a crude cell

    homogenate.

    Figure 7 shows the protein concentration and the glyox-

    alase II activity profiles obtained on the first oxidized

    glutathione-Sepharose affinity chromatographic column. All

    the glyoxalase I activity was eluted with the early

    fractions containing the bulk of the protein. Elution of

    glyoxalase II from the first column was accomplished by a

    gradient of the linear competitive inhibitor, S-octylgluta-

    thione. Before the addition of the pooled active fractions

  • H

    W A CQ < Eh

    H H

    W CO <

    J

    3

    o

    fa o

    25 O H Eh <

    U H fa H &

    D ft

    u 0 -P a H rd rH fa rd

    G CD O >

    •H O -P rd a

    •H MH •H ft U CD 3 •P ft CQ

    ' f t

    1—i 0) 0) cN> -P

    •rH '—' CO

    rd O > i 1 ^

    •H -p O) MH -H g •H > O *H • 1

    rd -P 0 -P

    -Prd •H ^ > •

    H ID

    a h

    G o

    •H •P a rd u fa

    O o • •

    rH CO ON r^

    CN CN

    CO

    CN

    If) G\

    CN

    o

    LO CD

    00

    CN CO • • •

    o o rH VD

    ro VO i n o VD

    rH rH rH

    o o o CO vo LO 00 VO

    G G G E 0 E

    •H rH 4-> H 0 rd 0 O

    a 1 rd i o ft a CO Q) CO CO U CO o ft o

    o

    a) -P G H3 CO 0

    o JH •H Q) a fa CO

    44 a 0

    •H -P rd

    -P G a) o G 0 u

    rd

    a> G 0

    •H £

    -P rd •P 3

    CO a) rd

    £ * > i i N >10 G -P g

    •H 0 > £

    •H •P i g o 0 -P v rd

    i—i l

    I 0 CO

    LO

    *0 (1) G

    •H rd •P G O a

    H -p

    X •H e

    •H -P a rd

    u •rH MH •H O i ft rd ft

    rd

    0) TJ •P (D MH

    > i O rd CO CO rd

    CO 0) rd

    H 0) •P rd e

    •H X 0 u ft ft rd

    CO •H

    rd

  • 45

    Figure 7

    First Oxidized Glutathione-Sepharose Affinity Chromatography

    of Swiss Mouse Liver Glyoxalase II

    Experimental details of chromatography and enzyme assays are

    described in Experimental Procedure. Crude prparation (380

    ml) was applied to a 2.6 x 70 cm preequilibrated oxidized

    glutathione-Sepharose column. Fraction volumes were 21 ml.

    The arrow marks the application of the S-octylglutathione

    gradient (1 to 5 mM) . o—o, glyoxalase II activity, -AA/min

    (75 pi of each fraction was used in the assay); • • ,

    protein concentration, mg/ml.

  • 46

    PROTEIN (mg/ml)

    - o

    e (uiui/VV)

    AJLIMJOV II aSVlVXOAlO

  • 47

    to the second affinity chromatographic column, the fractions

    were diluted two-fold to decrease the S-octylglutathione

    concentration. The reason for the dilution of the pooled

    active fractions was to allow binding of the enzyme to the

    second affinity column since binding is prevented by 2 mM

    S-octylglutathione. S-Octylglutathione also apparently

    stabilizes glyoxalase II and complete removal of this

    inhibitor by dialysis results in substantial loss of

    activity.

    Figure 8 shows the protein concentration and the glyox-

    alase II activity profiles obtained on the second oxidized

    glutathione-Sepharose affinity chromatographic column.

    Homogeneous glyoxalase II was eluted with 2 mM S-octylgluta-

    thione. The fractions which had high specific activities

    were pooled and concentrated by ultrafiltration.

    Although it is apparent that glyoxalase II binds to

    oxidized glutathione-Sepharose (second purification step),

    it is possible, because of the reduced glutathione present

    in the crude liver preparations (1-2 mM) used in the first

    purification step, that the Sepharose-bound oxidized

    glutathione can be reduced to reduced glutathione residues.

    Consequently, glyoxalase II binding could occur with both

    Sepharose-bound reduced and oxidized glutathiones. However,

    even after repeated use of the same affinity chromatographic

    column material with crude liver preparations, no change in

  • 48

    Figure 8

    Second Oxidized Glutathione-Sepharose Affinity

    Chromatography of Swiss Mouse

    Liver Glyoxalase II

    Experimental details of chromatography and enzyme assays are

    described in Experimental Procedure. Pooled and diluted

    fractions (920 ml) from the first column were applied to a

    2.6 x 40 cm preequilibrated oxidized glutathione-Sepharose

    column. Fraction volumes were 18.4 ml. The arrow marks the

    application of S-octylglutathione (2 mM). o — o , glyoxalase

    II activity, -AA/min (75 pi of each fraction was used in the

    assay); • — • , protein concentration, pg/ml.

  • 49

    P R O T E I N ( p g / m l )

    ( u i u i / w )

    A 1 I A I O V I I 3 S V 1 V X O J 1 1 9

  • 50

    glyoxalase II binding characteristics was observed. Further,

    pretreatment of the column material with several volumes of

    1 mM dithiothreitol does not change the glyoxalase II

    binding or elution patterns. After such usages or pre-

    treatments, an increase of Sepharose-bound sulfhydryl groups

    cannot be detected by use of Ellman's reagent (85). It

    would thus appear that the dithio group of Sepharose-bound

    oxidized glutathione is not readily reduced.

    Polyacryamide gel electrophoresis and sodium dodecyl

    sulfate polyacrylamide gel electrophoresis were conducted on

    the purified glyoxalase II. The amount of the enzyme

    applied to the gels varied from 8 to 20 pg protein. The

    gels exhibited only one band with both electrophoretic

    procedures; no minor bands could be detected either visually

    or by densitometric tracing of the gels.

    Molecular weight determinations. The weight-average

    molecular weight of native mouse liver glyoxalase I was

    estimated by the meniscus depletion method of sedimentation

    equilibrium ultracentrifugation. A plot of log(y-yQ)

    2

    versus r is shown in Figure 9. The linearity of the plot

    is additional evidence for the homogeneity of the enzyme

    preparation. The partial specific volume of glyoxalase I

    was taken as 0.728, determined from the partial specific

    volumes of the component amino acids (55). The molecular

    weight determined by sedimentation -equilibrium, 43,000

  • 51

    Figure 9

    Sedimentation Equilibrium Ultracentrifugation

    of Glyoxalase I

    Experimental details are given in Experimental Procedure.

    Ultracentrifugation was carried out at 14.4°c at 28,000

    rpm for 30 hours. The abscissa represents the square of the

    distance (cm) from the center of rotation. The ordinate

    represents the log of the fringe displacement in mm.

  • 52

  • 53

    daltons, agrees well with the value reported previously for

    the enzyme from the same source (55).

    Estimation of the molecular weight of mouse liver

    glyoxalase I by sodium dodecyl sulfate polyacrylamide gel

    electrophoresis using bovine serum albumin, ovalbumin and

    ribonuclease A as molecular weight standards, gave a value

    of 21,500 daltons (Figure 10). From the sedimentation

    equilibrium and sodium dodecyl sulfate polyacrylamide gel

    electrophoresis experiments, it has been concluded that

    mouse liver glyoxalase I has a molecular weight of approx-

    imately 43,000 and is a dimer composed of apparently

    identical subunits.

    A molecular weight estimation of mouse liver glyoxalase

    II was conducted by sodium dodecyl sulfate polyacrylamide

    gsl electrophoresis using ovalbumin, glyceraldehyde-

    3-phosphate dehydrogenase, triosephosphate isomerase and

    hemoglobin as molecular weight standards. The resulting

    experiments gave a value of approximately 29,500 daltons.

    Uotila estimated the molecular weight of human liver

    glyoxalase II to be 22,900 using gel filtration (57). In

    the light of this study, mouse liver glyoxalase II appears

    to be a single polypeptide with a molecular weight of

    approximately 29,500 (Figure 11)• Isoelectric focusing

    experiments were conducted on the homogeneous mouse liver

    glyoxalase II, and a pi value of approximately 8.1 was

  • 54

    Figure 10

    Molecular Weight Determination of Glyoxalase I by Sodium

    Dodecyl Sulfate Polyacrylamide Gel Electrophoresis

    Experimental details are given in Experimental Procedure.

    Molecular weight on a log scale versus relative distance of

    migration is plotted. Standards: Bovine serum albumin

    (BSA); ovalbumin (OVAL); ribonuclease A (RNase A). The

    migration of sodium dodecyl sulfate-treated glyoxalase I

    (G-I) indicates an apparent molecular weight of approx-

    imately 21,500 daltons.

  • 55

  • 56

    Figure 11

    Molecular Weight Determination of Glyoxalase II by Sodium

    Dodecyl Sulfate Polyacrylamide Gel Electrophoresis

    Experimental details are given in Experimental Procedure.

    Molecular weight on a log scale versus relative distance of

    migration is plotted. Standards: Ovalbumin (OVAL);

    glyceraldehyde-3-phosphate dehydrogenase (G3PDH); triose-

    phosphate isomerase (TPI); and hemoglobin (Hb). The

    migration of sodium dodecyl sulfate-treated glyoxalase II

    (G-II) indicates an apparent molecular weight of approx-

    imately 29,500 daltons.

  • 57

  • 58

    obtained. Uotila reported a pi value of 8.35 for the human

    liver enzyme (57).

    Inhibition of glyoxalase enzymes by nucleotides, nu-

    cleosides and a series of structurally related compounds.

    Nucleotides, nucleosides and some structurally related

    compounds are potent inhibitors of both glyoxalase enzymes

    derived from different sources (86). Figure 12 shows the

    activity response obtained for pure mouse liver glyoxalase I

    using GTP, ATP, CTP and UTP. Similar results (cooperative

    inhibitions) were obtained for pure mouse liver glyoxalase

    II. As an example, Figure 13 shows the effect of GTP on

    both enzymes; the inhibitions have almost identical

    sigmoidicities.

    S-Octylglutathione is a linear-competitive inhibitor of

    the glyoxalase enzymes, and it has been used as an affinity

    ligand for the purification of mouse liver glyoxalase I (54),

    as well as an eluant medium for mouse liver glyoxalase II

    purification from the affinity chromatography columns (58).

    The glyoxalase I inhibition response curve given by S-octyl-

    glutathione is quite different from the response curve given

    by nucleotides as shown in Figure 14 for GTP. When a Hill

    plot, log[%Inh./ (100- %Inh.)] versus log[GTP], (87) is

    constructed with the data of Figure 14, a Hill coefficient

    (slope) of 3.3 is obtained for the GTP inhibition (Figure

  • 59

    Figure 12

    Inhibition of Glyoxalase I by GTP, ATP, CTP and UTP

    The calculated hemimercaptal concentration is 0.2 mM. The

    free glutathione concentration, after equilibration, is 1.55

    mM. Glyoxalase I used in this study was the homogeneous

    enzyme preparation obtained from mouse liver (54). • •,

    GTP; • • , ATP; o o, CTP; • — • , UTP.

  • 60

    o> 80

    a>

    % 40 o a ^ 20

    100 200 300 400 500 [ILjjM

  • 61

    Figure 13

    Inhibition of Glyoxalase Enzymes by GTP

    The calculated hemimercaptal concentration is 0.2 mM. The

    free glutathione concentration, after equilibration, is 1.55

    mM. The S-£-lactoylglutathione concentration for the glyox-

    alase II assays is 0.4 mM. Both glyoxalases were obtained

    from mouse liver (54,58). • — g l y o x a l a s e I; A A ,

    glyoxalase II.

  • 62

    o> 80

    o O

    100 150 200 250 [GTPL/jM

  • 63

    Figure 14

    Inhibition of Glyoxalase I by GTP and

    S-Octylglutathione

    The calculated hemimercaptal concentration is 0.2 mM. The

    free glutathione concentration, after equilibration, is 1.55

    mM. The glyoxalase I used in this study was the homogeneous

    enzyme preparation obtained from mouse liver (54). • •,

    GTP; A A, S-octylglutathione.

  • 64

    100 150 200 250 [ILpM

  • 65

    Figure 15

    Hill Plot for the Inhibition of Glyoxalase I by GTP

    The data from Figure 14 were used to construct the plot. A

    value of 3.3 was obtained from the slope of the line.

  • 66

    c o

    I o o

    CD o

    1.2 1.4 1.6 13 2.0 log[colchicine]

  • 67

    15). In a binding study the Hill coefficient will theo-

    retically establish the minimum number of sites.

    Inhibitions of the mouse liver glyoxalase enzymes by

    some nucleic acid bases, nucleosides, nucleotides and a

    series of structurally related compounds are summarized in

    Table IV. 1 ^ values (the concentrations of the inhib-

    itors which give 50% inhibition) for each inhibitor are

    given; all compounds listed in the table inhibit both

    glyoxalase enzymes in a cooperative manner. Within a given

    family of nucleic acid bases, there is no difference in the

    extent of inhibitions given by the free base, the corre-

    sponding nucleoside, or the nucleoside 5'-mono-, di- or

    triphosphates. In addition to the inhibitors listed in

    Table IV, pyridine nucleotides, cAMP and cGMP were also

    studied. Significant inhibitions (35-60%) of both glyox-

    alase enzymes were given by these nucleotides at 150 pM

    concentrations. In general, aromatic compounds which

    contain heterocyclic nitrogen and/or have amino groups on

    the aromatic ring, are effective cooperative inhibitors of

    both glyoxalase enzymes. Compounds not having these

    structural features (e.g., tyrosine, phenylalanine, histi-

    dine, arginine, imidazole) do not inhibit either enzyme,

    even at concentrations above 2 mM.

    Comparisons of the inhibition by nucleotides of both

    glyoxalase enzymes from different sources were undertaken.

  • > H

    w J

    CQ <

    E h

    CO

    o s

    o

    E h

    H

    CQ

    H

    £

    2 5

    W

    CO

    C J

    o

    t H

    h q

    0

    w

    s

    o

    CO

    o

    3 .

    CO

    w

    ID

    0 o

    L O

    H

    • P

    G a ) r o ( N

    • H • •

    h a CO CO

    H - H I I I I • H m P ^ ft

    MH EH EH

    i

    H

    0

    u o

    - p

    • H

    • Q

    • H

    G

    0 0 •

    CM I I

    PL, E h

    D

    o m o o o o o m o o o o m o o

    v o r ^ i n m < r k i n o r ^ o i n i n i n r - o o

    r H r H C N C N C N r O L O ^ L D L n L O L T ) L O v D C 0

    o m o m o o m o o o o i n o o o

    L O M ^ h O i D h O O m i n h O O O

    r H r H C N i c N f o o o ^ L n L o m i n i n v o v o c ^

  • 69

    There are no significant differences between the inhibition

    of mouse liver glyoxalase I and the yeast enzyme. Nucleo-

    tides are comparatively better inhibitors of the glyoxalases

    from Pseudomonas fluorescens than of these enzymes from

    mouse liver. Interestingly, the inhibition of glyoxalase II

    from the bacterial source does not exhibit sigmoidal

    kinetics.

    Since intracellular levels of some of the nucleotides

    (88, 89), for example ATP, are significantly higher than the

    concentrations needed to inhibit both enzymes completely

    (0.5 mM), the inhibitions observed may have physiological

    significance. The glyoxalases are either completely in-

    active in vivo (there is experimental evidence to the

    contrary (90)), or they are active because of isolation from

    inhibitors through compartmentalization. An alternate ex-

    planation is that there is a variable endogeneous activator

    which reverses the inhibition of the enzymes, thereby

    regulating glyoxalase activities. The identification of

    such an activator would provide much information about the

    function of the glyoxalase enzymes.

    Inhibition of Glyoxalase Enzymes by Microtubule Poisons.

    As previously mentioned, it was shown that nucleotides,

    nucleosides and some structurally related compounds were

    potent, cooperative inhibitors of both glyoxalase enzymes

    (86). The biological significance of these inhibitions is

  • 70

    not readily apparent. It was found that certain microtubule

    poisons (colchicine, vinblastine, podophyllotoxin and

    griseofulvin) also inhibit both glyoxalases in a cooperative

    manner. As an example, in Figure 16, the inhibitions of

    homogeneous mouse liver glyoxalase I and glyoxalase II by

    colchicine are shown, and the inhibitions have almost

    identical sigmoidicities. When a Hill plot is constructed

    for the glyoxalase I response curve (Figure 17), a value of

    3.7 (from the slope) was obtained for the Hill coefficient.

    The Hill coefficients were also similarly determined for

    glyoxalase I inhibitions by vinblastine, podophyllotoxin and

    griseofulvin, and are 3.6, 3.4 and 3.1, respectively.

    Figure 18 shows the Lineweaver-Burk plot for the inhibition

    of glyoxalase I by colchicine. A Dixon plot of the data

    indicated that colchicine is a nonlinear-competitive

    inhibitor. Plots derived from data of glyoxalase II inhi-

    toition toy colchicine are quite similar to those shown in

    Figures 17 and 18 for glyoxalase I inhibition.

    After the observation of the inhibition of glyoxalases

    by the microtubule poisons, the questions arose: What is the

    specificity of the inhibitions observed? Are these micro-

    tubule poisons general enzyme inhibitors? As an approach to

    answering these questions, the effects of colchicine,

    vinblastine, podophyllotoxin and griseofulvin on various

    enzymes from a variety of sources were examined, and Table V

  • 71

    Figure 16

    Inhibition of Glyoxalase Enzymes by Colchicine

    The calculated hemimercaptal concentration is 0.2 mM. The

    free glutathione concentration, after equilibration, is 1.55

    mM. The S-D-lactoylglutathione concentration for the glyox-

    alase II assays is 0.4 mM. Both glyoxalases were obtained

    from mouse liver (54,58). • •, Glyoxalase I; o o,

    glyoxalase II.

  • 72

    o> c 80 c o £ CD 60 >*

    • a m >

  • 73

    Figure 17

    Hill Plot for the Inhibition of Glyoxalase I by Colchi cine

    The data from Figure 16 was used to construct the plot. A

    value of 3.7 was obtained from the slope of the line.

  • 74

    ' 0.5

    • a - 0 5 o

    12 1.4 1.6 15 log[colchicine]

  • 75

    Figure 18

    Lineweaver-Burk Plot of the Effect of Colchicine

    Concentration on the Glyoxalase I Activity

    The assay method is described in Experimental Procedure.

    Velocity is in terms of pioles of S-g-lactoylglutathione

    produced per minute. The free glutathione concentration,

    after equilibration, is 1.55 mM, for each point. The

    hemimercaptal concentration was varied with the following

    constant levels of colchicine: 0 (•), 30 jaM (•), 40 pM (A),

    45 JJM (o) , 50 JAM (•) .

  • 76

    3 30 > s

    2 4 6 8 10 12 1 / I Hemimercaptall.mM

  • w a CQ < Eh

    CO W s

    £S3 £ w

    CO p o H &

    g

    53 O

    co £ O CO H O (X

    w J p ca D

    O & u

    fa o Eh O W fa fa M

    >i X)

    G 0 •H •P •H .0 •H £ G

    o\P

    2 a

    o LO

    s 3u

    O ^0

    G •H >

    2 H a 3

    m o in

    a> g

    "H s o OL-H

    0 * 0 LO H

    0 U

    i N G W

    o o o o lo in m

    o o o o in in in

    o o o o in in in

    o o o o in m in

    a) CO rd G a) cn

    — 0 u u

    }H >i > •H £

    •H rH 0 H

    0 a) 4J CO 0 CO CO 0 -P

    rd 0 rd 0 a) E & g >i

    CO H 0

    H M H rG cu

    i >i >i 3 rH rH rH rH o o O 0

    OCNO

    o o o

    o o o

    o o o

    -p CO rd 0 >1

    a> CO rd g 0 tn 0 u nd >i

    0 0 CO *3 •H

    0 •P 0 -P CO -P rd U 0 U rd rd £ co -P co 0 £ On 3 rd 3 >i & co g £ g - W O On

    0 £ -P CO -P ,G (1|-H 0 -H

    SH rQ d> rd 0 M > i w rH 0 1 2$

    0

    q a • •

    S3 53

    53

    o o o

    a>

    CD (0 o u

    rH o

    0 CO 0 •H $-1 Eh

    0 rH U CO 3 g

    -P H

    0 rH a co 3 g

    -P •H £ £ rd Jh

    O O C N O O rH

    o o o o o o o o o

    o o o o o o

    o o o o o o

    c 0 a i

    rH >1 u — rd u -p 0 3 > rH -H CP rH H >1 -p

    Q) CO rd

    S * 3 u rd •P G rd

    •P G >i co

    n3 •H a rd

    A * rQ 0 rd -P

    a) mh CO 0 rd rC

    O CO 3

    0 co rd

    >i -P X U 0 3 u

    CUTQ Q) CO >1 u O EC rC I a» oo

    J l -P g — +> rd

    0 MH CO rd a) •P g 0 rd rC 04 04 -P o G Jh • >i Qii-M CO 0 —'

    2 U 2 >i co a

    CO H M rd

    0 O CO CO rd

  • 78

    summarizes the results# The levels of these drugs tested

    are the concentrations which gave 50% inhibition (I 50

    values) for the glyoxalase enzymes. It was found that only

    the glyoxalases were inhibited by these microtubule poisons,

    and that no significant inhibition could be detected for any

    of the other enzymes investigated at the inhibitor

    concentrations employed.

    Tubulin is known to have two binding sites for guano-

    sine nucleotides (64) and tubulin also binds the drugs

    tested. It is therefore, interesting to note that some of

    the compounds known to bind tubulin and/or affect micro-

    tubule assembly also inhibit both glyoxalase enzymes. It is

    also known that lumicolchicine does not bind tubulin (91);

    therefore, experiments were conducted to determine the

    effects of lumicolchicine on the glyoxalase enzymes. As

    shown in Figure 19, the inhibition of glyoxalase I by

    lumicolchicine is not sigmoidal, and significantly less

    inhibitory activity is exhibited by it as compared to

    colchicine. It is apparent that lumicolchicine binds to a

    noncatalytic site on dimeric glyoxalase I (54), producing an

    altered enzyme, since a maximum of only 50% inhibition is

    obtained. Lumicolchicine, on the other hand, does not

    inhibit glyoxalase II to any appreciable extent.

    On the basis of the inhibitions exhibited by micro-

    tubule poisons on glyoxalase I and glyoxalase II, it is

  • 79

    Figure 19

    Inhibition of Glyoxalase Enzymes by Colchicine

    and Lumicolchicine

    The calculated hemimercaptal concentration for glyoxalase I

    assays is 0.2 mM. The free glutathione concentration, after

    ®ciuilibration, is 1.55 mM. The S—D— lactoylglutathione

    concentration for glyoxalase II assays is 0.4 mM. m • ,

    inhibition of glyoxalase I or glyoxalase II by colchicine;

    • •, inhibition of glyoxalase I by lumicolchicine;

    the effect of lumicolchicine on glyoxalase II.

  • 80

    !> 40

    20 40 60 80 [Colchicine or LumicolchicineLjjM

  • 81

    tempting to speculate that these two enzymes may have some

    involvement in the microtubule assembly process. Such spec-

    ulations have appeared in the literature (60-62). It has

    been suggested that the thioester, S-D-lactoylglutathione

    (the product of glyoxalase I and the substrate of glyoxalase

    II), may have a significant role in the formation of the

    mitotic apparatus of dividing eucaryotic cells (62). In

    relation to the postulate that a-ketoaldehydes have a

    special function as regulatory molecules in cell division

    (32,33,35), concanavalin A, a cell-division stimulator, has

    been shown to increase the activity of both glyoxalase

    enzymes in lymphocytes and polymorphonuclear leukocytes

    (61). It is apparent that the intracellular concentrations

    of both glutathione and S-D-lactoylglutathione could well be

    affected by such an activation process. There is also

    evidence in the literature that the glyoxalase system may

    regulate microtubule assembly in vitro (60). The effect of

    low levels of S-D-lactoylglutathione on the assembly of

    purified tubulin was determined, and it was found in a

    number of experiments to increase the rate of microtubule

    assembly. In addition, it has been demonstrated (62) that

    S-D-lactoylglutathione enhances the rate of anti-IgE-induced

    histamine release from human leukocytes - a secretory

    process believed to require an intact microtubule assembly

    (92).

  • 82

    In light of this study, it is concluded that the rather

    specific inhibitions of the glyoxalases by microtubule

    poisons may have metabolic significance. However, the

    evidence for a proposed functional relationship between the

    glyoxalase enzymes and microtubule assembly is largely

    circumstantial at this point. Further investigations in

    this direction may bring into light the biological function

    of glyoxalases and a better understanding of the regulation

    of microtubule assembly-disassembly processes.

    Kinetics. Excessive free glutathione concentrations

    were found to be inhibitory to mouse liver glyoxalase I

    activity (23). Mannervik and coworkers (24) have subjected

    several steady-state kinetic models to linear and nonlinear

    regression analyses. It was concluded that the kinetics of

    glyoxalase I are best described by a model including alter-

    nate one- or two-substrate pathways. Except at very low

    hemimercaptal and free glutathione concentrations, the one-

    substrate pathway predominates. At higher hemimercaptal and

    free glutathione concentrations the one-substrate pathway

    model predicts a hemimercaptal-free glutathione competition.

    The effects of varying concentrations of free glutathione on

    the reaction kinetics of glyoxalase I were studied. As

    shown in Figure 20 free glutathione was indeed a competitive

    inhibitor. This effect has been previously investigated

    (93), and it was concluded that free glutathione was

  • 83

    Figure 20

    Lineweaver-Burk Plot of the Effect of Glutathione

    Concentration on the Glyoxalase I Activity

    The assay method is described in Experimental Procedure.

    Velocity is in terms of jamoles of S-D-lactoylglutathione

    produced per minute. The hemimercaptal concentration was

    varied with the following constant levels of free gluta-

    thione: 0.3 mM (•), 3.0 mM (•), 6.0 mM (A), 8.0 mM (o),10.0

    mM (A) .

  • 84

    2 4 6 8 10 12

    1 / [ H e m i m e r c a p t a l ] , m M

  • 85

    essentially competitive with the hemimercaptal for mouse

    liver glyoxalase I.

    S-Octylglutathione, which was used as the affinity

    ligand in the purification of mouse liver glyoxalase I, was

    shown to be a potent inhibitor of glyoxalase I (41).

    Examination of the Lineweaver-Burk plot shown in Figure 21

    reveals that S-octylglutathione is a linear-competitive

    inhibitor of glyoxalase I. By using the graphical method of

    Dixon (94), the for S-octylglutathione was determined

    to be 0.06 mM.

    Experiments were carried out with varying free gluta-

    thione and S-octylglutathione concentrations at constant

    levels of hemimercaptal to determine whether kinetically

    distinguishable binding sites are available simultaneously

    for glutathione and S-octylglutathione. The parallel lines

    obtained in the plot of l/v versus S-octylglutathione

    concentrations (Figure 22) indicate that these two compounds

    are mutually exclusive as effectors of the enzymatic activ-

    ity (95). The fact that glutathione and its S-substituted

    derivative are not only mutually exclusive, but also indi-

    vidually compete with the hemimercaptal, suggests that the

    site of binding is the active site of glyoxalase I. Similar

    experiments were conducted to determine the effects of

    various fixed concentrations of S-octylglutathione on the

    glyoxalase I activity at increasing levels of free

  • 86

    Figure 21

    Lineweaver-Burk Plot of the Effect of S-Octylglutathione

    Concentration on the Glyoxalase I Activity

    The assay method is described in Experimental Procedure.

    Velocity is in terms of pmoles of S-p-lactoylglutathione

    produced per minute. The hemimercaptal concentration was

    varied with the following constant levels of S-octylglu-

    tathione: 0 (•), 50 pM (•), 100 pM (A), 150 pM (o), 200 pM

    (•).

  • 87

    l/[Hemimercaptal],mM

  • 88

    Figure 22

    Effect of Various Fixed Concentrations of S-Octylglutathione

    on the Glyoxalase I Activity at Increasing Levels of Free

    Glutathione with a Constant Level of Hemimercaptal

    The assay method is described in Experimental Procedure.

    Velocity is in terms of pmoles of S-D-lactoylglutathione

    produced per minute. The hemimercaptal concentration was

    constant at 0.3 mM for all points. S-Octylglutathione

    concentrations were: 0 (•), 25 pM (•), 50 pM (A), 75 pM (o),

    100 >iM (A).

  • 89

    [GSHf],mM

  • 90

    methylglyoxal in the presence of a constant level of hemi-

    mercaptal. From the plot of l/v versus free methylglyoxal

    concentration (Figure 23), which results in parallel lines,

    it is concluded that these two compounds are mutually

    exclusive as effectors of the enzymatic activity.

    As shown in Figure 24, parallel lines were obtained

    when l/v versus colchicine concentration were plotted with

    varying levels of S-octylglutathione in the presence of a

    constant level of hemimercaptal. This indicates that col-

    chicine is also mutually exclusive with S-octylglutathione.

    Glutathione, methylglyoxal, S-octylglutathione and

    colchicine appear to bind the active site of glyoxalase I,

    and this conclusion can be substantiated by the observations

    that both glutathione and S-octylglutathione compete with

    the hemimercaptal, and that both free methylglyoxal and

    colchicine are mutually exclusive with respect to S-octyl-

    glutathione. When free glutathione is tested with various

    fixed concentrations of colchicine at a constant level of

    hemimercaptal (Figure 25), the lines are not parallel, but

    they cross on the vertical axis. The first interpretation

    of the data would be that colchicine and S-octylglutathione

    are not mutually exclusive. When two inhibitors can bind to

    the same enzyme molecule (when they are not mutually

    exclusive) the lines would be expected to cross somewhere to

    the left of the vertical axis (96). One way of interpreting

  • 91

    Figure 23

    Effect of Various Fixed Concentrations of S-Octylglutathione

    on the Glyoxalase I Activity at Increasing Levels of Free

    Methylglyoxal with a Constant Level of Hemimercaptal

    The assay method is described in Experimental Procedure.

    Velocity is in terms of pinoles of S-D-lactoylglutathione

    produced per minute. The hemimercaptal concentration was

    constant at 0.3 mM for all points. S-Octylglutathione

    concentrations were: 0 (•) , 25 jjM (A), 50 pM (•), 100 pM

    (A).

  • 92

    2 £

    © S

    A/I

  • 93

    Figure 24

    Effect of Various Fixed Concentrations of S-Octylglutathione

    on the Glyoxalase I Activity at Increasing Levels of

    Colchicine with a Constant Level of Hemimercaptal

    The assay method is described in Experimental Procedure.

    Velocity is in terms of pmoles of S-g-lactoylglutathione

    produced per minute. The hemimercaptal concentration was

    constant at 0.3 mM for all points. S-Octylglutathione

    concentrations were: 0 (•) , 25 juM (•) , 50 pM (A), 75 pM (o) ,

    100 pM (•).

  • 94

    [Colchicine] , j jM

  • 95

    Figure 25

    Effect of Various Fixed Concentrations of Colchicine on

    the Glyoxalase I Activity at Increasing Levels of Free

    Glutathione with a Constant Level of Hemimercaptal

    The assay method is described in Experimental Procedure.

    Velocit