throbber
Neurochemical Research, Vol. 16, No. 9, 1991, pp. 965-974
`
`An Overview of 'Y-Hydroxybutyrate Catabolism: The Role
`of the Cytosolic NADP+-Dependent Oxidoreductase EC
`1.1.1.19 and of a Mitochondrial Hydroxyacid-Oxoacid
`Trans hydrogenase in the Initial, Rate-Limiting Step in
`This Pathway
`
`Elaine E. Kaufrnan1 and Thomas Nelson1
`
`(Accepted April 22, 1991)
`
`'Y-Hydroxybutyrate (GHB) is a naturally occurring
`compound present in micromolar concentration in both
`brain (1,2) and in peripheral tissues (3). This endoge(cid:173)
`nous compound is remarkable in that pharmacological
`doses of 200-500 mg/kg produce marked behavioral and
`electroencephalographic changes (4), a profound de(cid:173)
`crease in cerebral glucose utilization (5), an increase in
`striatal dopamine levels ( 6) and a decrease in body tem(cid:173)
`perature (7). High doses of GHB have also been reported
`to protect neurons (8) and intestinal epithelium (9) against
`cell death resulting from experimental ischemia. Behav(cid:173)
`ioral changes are not seen with doses of less than 30 mg/
`kg, but low doses stimulate the release of prolactin, growth
`hormone and cortisol (10,11), and doses of 5-10 mg!kg
`result in an increase in body temperature (12). These
`actions, and the discovery of high affinity binding sites
`for GHB in the central nervous system (13), suggest that
`GHB may have a biological function. Both the origin of
`endogenous GHB and its catabolism are, therefore, of
`considerable interest.
`This review will cover the early work on the deg(cid:173)
`radative pathway for GHB and the discovery of a dual
`pathway for the initial step in the oxidative catabolism
`of GHB. The factors which regulate the activity of the
`
`1 Laboratory of Cerebral Metabolism, National Institute of Mental Health,
`United States Public Health Service, Department of Health and Hu(cid:173)
`man Services, Building 36, Room lA-05, Bethesda, Maryland 20892
`* Special issue dedicated to Dr. Louis Sokoloff.
`Abbreviations used in this paper: GHB, ')'·hydroxybutyrate; SSA,
`succinic semialdehyde; DTI, dithiothreitol.
`
`enzymes in these pathways, and as a consequence, reg(cid:173)
`ulate tissue levels of GHB are also discussed.
`Walkenstein et al. (14) established that GHB is
`largely disposed of, in vivo by oxidation to C02 and
`water. These investigators could not find the 14C label
`from GHB in succinate in urine, but they did find that
`the label could be trapped in hippuric acid in the urine
`of animals treated with sodium benzoate as might be
`expected if GHB were undergoing 13-oxidation. They
`therefore proposed that GHB was metabolized by 13-ox(cid:173)
`idation (14). Mohler et al. (15) and Doherty et al. (16),
`however, assayed citric acid cycle intermediates in the
`tissues of animals given [14C]GHB and demonstrated
`that the carbon skeleton of GHB indeed does enter the
`citric acid cycle as succinate rather than as acetyl-CoA
`as would be expected if GHB were being oxidized through
`the 13-oxidation pathway.
`They proposed the following pathway:
`1) GHB ~ succinic semialdehyde
`2) Succinic semialdehyde --7 succinate
`3) Succinate ~ ~ ~ ~ Co2 + H20
`The discovery of a metabolic disease in which GHB
`and succinic semialdehyde (SSA) are markedly elevated
`(17) in both blood and urine due to a block in SSA
`dehydrogenase (18) added evidence to support a degra(cid:173)
`dative pathway in which GHB is oxidized to SSA, which
`in turn is oxidized to succinate.
`At the time the pathway shown above was pro(cid:173)
`posed, it was known that the enzymes of the citric acid
`cycle catalyzed the reactions in step (3) and that SSA
`
`965
`
`0364-3190/91/0900-0965$06.50/0 © 1991 Plenum Publishing Corporation
`
`Ranbaxy Ex. 1017
`IPR Petition - USP 9,050,302
`
`

`
`966
`
`Kaufman and Nelson
`
`dehydrogenase (the enzyme missing in patients with OHB
`aciduria (18)) catalyzed the reaction in step (2). Step (1)
`is always depicted as an essential part of this scheme.
`However, at the time this pathway was proposed, an
`enzyme or enzymes which could catalyze this step had
`not been identified.
`Isolation of a Cytosolic GHB Dehydrogenase. Two
`unusual oxidoreductases, one cytosolic (19) and the other
`mitochondrial (20), that catalyze the oxidation of OHB
`to SSA have now been isolated. The cytosolic enzyme,
`which will be referred to as ORB-dehydrogenase in this
`review, was first purified to homogeneity from hamster
`liver (19) and was found to be an NADP+-dependent
`oxidoreductase. A study of the substrate specificity of
`purified ORB-dehydrogenase revealed that D-glucuron(cid:173)
`ate and L~gulonate, the product of D-glucuronate reduc(cid:173)
`tion, were also good substrates (19). The physical
`characteristics, as well as the substrate and inhibitor
`specificity of this enzyme, indicate that the ability to
`catalyze the oxidation of OHB probably represents a pre(cid:173)
`viously unreported activity for the NADP+-dependent
`oxidoreductase (EC 1.1.1.19) commonly known as D(cid:173)
`glucuronte reductase (21). This enzyme may also be
`identical to the group of enzymes categorized in a 1985
`review by Cromlish et al. (22) as "ALR-1", the high
`Km aldehyde reductase or L-hexonate dehydrogenase.
`Although the oxidation of OHB catalyzed by this
`ORB-dehydrogenase proceeds at an easily measurable
`rate when assayed in vitro under optimal conditions, the
`very low activity found in the in vitro system under
`conditions simulating those in the cytosol raises the
`question of how, or indeed whether, this enzyme cata(cid:173)
`lyzes the oxidation of OHB in vivo. An answer to this
`question may have been found when it was discovered
`that OHB dehydrogenase could catalyze the reduction of
`o-glucuronate coupled to the oxidation of GHB (23) as
`shown below:
`OHB + NADP+ :;;:= SSA + NADPH + H+
`o-glucuronate + NADPH + H + :;;:= L-gulonte +
`NADP+
`The overall or "coupled" reaction is:
`GHB + o-glucuronate :;;:= SSA + L-gulonate
`When the kinetic constants for the coupled system were
`determined, it was found that they were more favorable
`to oxidation of GHB under conditions present in cytosol
`of most tissues th~n were those for the uncoupled sys(cid:173)
`tem.
`
`The time course of the coupled reaction (Figure 1)
`in which both products, SSA and NADPH, were mea(cid:173)
`sured, shows that in the presence of an adequate con(cid:173)
`centration of o-glucuronate, only a very small net amount
`of NADPH is formed even though SSA formation is
`
`. 10
`
`. 08
`
`i . 06
`
`"' :!l
`.
`';;
`" . 04
`E ,_
`
`0
`
`. 02
`
`0
`
`0
`
`SSR
`
`""
`
`NRDPH
`
`I
`
`10
`
`12
`
`14
`
`16
`
`18
`
`20
`
`Time
`( m 1 n)
`Fig. 1. Time course of succinic semialdehyde (SSA) and NADPH
`formation in the presence of D-glucuronate (23). The reaction mixture
`contains 10 mM GHB, 1.0 mM D-glucuronate, 0.1 mM NADP+, 80
`mM phosphate buffer, pH 7.6, enzyme and sufficient water to bring
`the volume to 1.0 mi. SSA and NADPH were determined as previously
`described (19).
`
`proceeding rapidly. This is in contrast to the control
`reaction mixture without glucuronate in which NADPH
`and SSA are formed in stoichiometric amounts (19). These
`results indicate that NADPH is being used for the re(cid:173)
`duction of D-glucuronate at the same rate at which it is
`being produced by the oxidation of GHB. This would
`account for the low steady state level of NADPH seen
`in Figure 1. The effect of increasing concentrations of
`D-glucuronate on the rate of oxidation of GHB to SSA
`in the presence of limiting amounts of NADP+ and in(cid:173)
`hibitory amounts of NADPH is shown in Figure 2. Un(cid:173)
`der these conditions, 2 mM o-glucuronate increased SSA
`formation 8-fold.
`Other important changes in the kinetic constants for
`this reaction occur in the presence of D-glucuronate (Ta(cid:173)
`ble 1). The Km (4.5 x 10- 4M) for the coupled reaction
`is fiVe-fold lower and the V max (1.52 J.Lmol/min/mg pro(cid:173)
`tein) is 1.8 times higher than in the uncoupled reaction.
`The effects of coupling and changes in pH on the rate
`of degradation of OHB are, however, more accurately
`described by changes in k (the first order rate constant
`for the reaction) than they are by changes in V max' The
`concentration of GHB in the tissues is much lower than
`Km and under these conditions the quantity V max!Km pro(cid:173)
`vides a good approximation of k (24). In the coupled
`reaction, the rate constant for GHB degradation, V maxi
`Km, is increased 9-fold as compared to the 1.8-fold in(cid:173)
`crease in V max at saturating concentrations of OHB and
`NADP+ (Table 1). The effect of o-glucuronate on the
`rate of the reaction is much greater at the very low sub-
`
`

`
`Enzymes Which Catalyze the Initial Step in GHB Catabolism
`
`967
`
`. 40
`
`.35
`
`~ .30
`
`~ .25
`' d:
`ill
`~ .15
`
`. 20
`
`. 10
`
`.05
`
`.4
`
`. 8
`
`I. 2
`
`!.&
`
`[0-Glucuronate) (mM)
`
`Fig. 2. The effect of D-glucuronate concentration on the rate of con(cid:173)
`version of GHB to succinic semialdehyde (SSA) in a reaction mix
`containing NADPH and a low concentration of NADP+ (23). Reaction
`mixture: 10 mM GHB, 0.01 mM NADP+, 0.02 mM NADPH, 80 mM
`phosphate, pH 7.6, enzyme, o-glucuronate as indicated and water to
`1 mi. The assay for SSA was carried out as previously described (19).
`
`strate concentrations found in vivo than in the standard
`in vitro assay where V max conditions are used. In the
`coupled reaction the Km for NADP+ is decreased from
`2 x l0- 5 to 1.4 x 10-6M and the inhibition produced
`by NADPH (Ki = 7 x 10- 6M) has been eliminated
`(Figure 3). GHB can now be oxidized in the presence
`of an otherwise extremely inhibitory concentration of
`NADPH. All of the kinetic constants· for the coupled
`reaction are closer to the tissue concentration range shown
`in Table I (25) than are those for the uncoupled reaction.
`The rate of GHB oxidation is pH-dependent. Earlier
`work had shown that the pH optimum for the cytosolic
`oxido-reductase under V tnax conditions was 9.0 (19), but
`
`at a more physiological pH (7.0-7.2) the enzyme was
`only half as active. The pH optimum was dependent on
`GHB concentration in both the coupled and uncoupled
`reactions and in both cases was above 8.0 when satu(cid:173)
`rating concentrations of substrate were used (26). As the
`concentration of GHB decreases toward levels found in
`vivo the pH optimum for the coupled reaction shifts toward
`pH 7.0 (26). A plot of V max!Krn against pH (Figure 4)
`shows that, at substrate concentrations near those found
`in vivo, the pH optimum approaches the intracellular
`pH, i.e. 7.5 for the uncoupled reaction and 7.0 or lower
`for the coupled reaction (26). Vayer et al. (27) subse(cid:173)
`quently reported a pH optimum of 8.0 under different
`conditions from those described above. In their system
`the oxidation of GHB catalyzed by the cytosolic oxi(cid:173)
`doreductase was coupled to both the reduction of D(cid:173)
`glucuronate and the transamination of SSA to form GABA.
`GHB dehydrogenase is inhibited by a number of
`products of intermediary metabolism (Table II) which
`includes the ketone bodies, a'.-ketoglutarate and branched
`0'.-ketoacids derived from amino acid degradation as well
`as degradation products of phenylalanine (26). As has
`been found with certain aldehyde reductases (28,29),
`anticonvulsants such as barbiturates, diphenylhydantoin
`and valproate are good inhibitors of GHB dehydrogenase
`(30). In addition, GHB dehydrogenase is inhibited by
`salicylates (30)-
`GHB dehydrogenase like lysozyme, ribonuclease
`and a number of other proteins (26), may contain disul(cid:173)
`fide bridges which are essential for its activity. It is
`inhibited by compounds such as 13-mercaptoethanol and
`dithiothreitol (DTI), which can reduce disulfide bonds
`(26). DTT has the most pronounced effect; addition of
`2.5 mM DTT produces an 85% inhibition of the activity
`
`Table I. The Effect of D-Glucuronate on the Kinetic Constants for -y-Hydroxybutyrate (GHB), NADP+, and NADPH
`
`GHB
`NADP+
`NADPH
`GHB
`V m,./K,GHB
`
`Kinetic Constants
`
`Uncoupled assay
`
`2.3 X 10·3 M
`2 X 10-5 M
`7 X 10-6 M
`0.83
`0.36
`
`Coupled assay
`
`4.5 X lQ-<Mh
`1.4 X 10-6 M<
`No inhibition
`1.52"
`3.38
`
`Tissue concentration
`
`(range between brain,
`liver, kidney, muscle)"
`0.1-5 X 1Q-S M
`2-11 X 10" 6 M
`1-30 X 10-5 M
`
`"The tissue concentrations of GHB are from reference (3). The molar concentration of NADP+ and NADPH in the various tissues were calculated
`from data taken from reference (25).
`•1 mM D-glucuronate.
`<2 mM D-glucuronate.
`dlJ.mol/min/mg protein.
`•first order rate constant when [S] < < Km
`Data in this table are from reference (23)
`
`

`
`+NADPH
`
`Table II. Kinetic Constants for the Inhibition of GHB(cid:173)
`Dehydrogenase (26,30)
`
`Kaufman and Nelson
`
`CONTROL
`
`968
`
`"-
`
`E
`
`'
`~
`IS
`!!' 1/v
`
`~
`
`ill
`6
`
`-E
`
`
`"-
`
`Inhibitor
`
`DL-13-Hydroxybutyrate
`Acetoacetate
`a-Ketoglutarate
`p-Hydroxyphenylpyruvate
`Phenylacetate
`Pyruvate
`a-Ketoisovalerate
`a-Ketoisocaproate
`DL-a-keto-13-methyl n-Valerate
`Valproate
`Salicylate
`
`K; (mM)
`
`Uncoupled
`reaction
`
`Coupled
`reaction
`
`4.4
`
`0.6
`
`0.4
`
`0.2
`
`3.9
`3.0
`1.1
`1.0
`0.5
`7.0
`0.33
`0.17
`0.06"
`0.057
`0.115
`
`Assays for the coupled reaction and the uncoupled reaction were car(cid:173)
`ried out as described (26).
`"It should be noted that the K; has only been reported for the DL(cid:173)
`mixture.
`
`not been previously inhibited by sulfhydryl reducing
`agents.
`When the activity of GHB dehydrogenase is being
`measured, the inclusion of reducing compounds such as
`13-mercaptoethanol or DTT in the assay mixture as has
`been reported by some laboratories (27) could lead to
`incorrect results.
`Identification of a Mitochondrial Transhydrogenase
`Which Catalyzes the a-Ketoglutarate-Dependent Oxi(cid:173)
`dation of GHB. The discovery of GHB dehydrogenase
`made it possible to complete a sequence of reactions
`leading from GHB to C02 and H20. Development of a
`polyclonal antibody to the GHB dehydrogenase then made
`it possible to determine whether or not there were ad(cid:173)
`ditional enzymes in the cytosol or in other subcellular
`fractions that could catalyze the initial, and probably
`rate-limiting, step in the oxidation of GHB to C02 (31).
`The antibody, which inhibited ""' 95% of the ability of
`the cytosolic fraction to oxidize GHB to SSA, failed to
`inhibit approximately 60% of this activity in kidney ho(cid:173)
`mogenate (Figure 5). These results suggested that there
`was at least one additional enzyme which catalyzed the
`conversion of GHB to SSA. The mitochondrial fraction,
`which had previously been shown to lack GHB dehy(cid:173)
`drogenase activity (19), could oxidize [14C]GHB to 14C02
`and could catalyze the pyridine nucleotide-independent
`oxidation of GHB to SSA (31). The mitochondrial en(cid:173)
`zyme was subsequently isolated and shown to be a hy(cid:173)
`droxyacid-oxoacid transhydrogenase which catalyzed the
`following reaction (20):
`GHB + a-ketoglutarate ~ SSA + a-hydroxyglutarate.
`
`18
`
`20
`
`30
`
`40
`
`50
`
`Fig. 3. The effect of D-glucuronate on the kinetics of the oxidation
`of GHB with NADP+ as the variable substrate (23). In the absence of
`D-glucuronate, NADPH formation was measured; in the presence of
`D-glucuronate, succinic semialdehyde (SSA) formation was measured.
`• = control, • = control with 0.02 mM NADPH, o = control with
`2.0 mM D-glucuronate, o = control with 0.02 mM NADPH and 2.0
`mM D-glucuronate.
`
`A.
`
`vmax , Krn
`
`0.24
`
`0.22
`
`coupled
`
`0.20
`
`- .::
`E
`y: ~ 0.18
`~ .s 0.16
`E ~
`>
`
`0.14
`
`0.12
`
`0.10
`7.0
`
`uncoupled
`
`7.5
`pH
`
`8.0
`
`E
`~ 0.8
`E
`
`0 :f'-~/\
`~ 2E 0.6 >if
`OOL_j
`
`'
`
`I
`
`8
`pH
`
`10
`
`Fig. 4. The effect of pH on the ratio of V mflm and on V mox for the
`oxidation of GHB by GHB dehydrogenase. The data for Figure 4A
`were taken from reference (26). The data in Figure 4B are from ref(cid:173)
`erence (19). VmJ~ = the first order rate constant, k, when [S] < <
`Km•
`
`of the purified enzyme and 2.0 mM DIT inhibits 84%
`of the activity found in rat liver cytosol. The inhibition
`produced by DTT can be partially reversed by the ad(cid:173)
`dition of H20 2 or completely reversed by oxidized glu(cid:173)
`tathione. Preincubation of GHB dehydrogenase with 2.5
`mM DTT before it is added to the reaction mixture (and
`thereby diluted 50 fold) does not inhibit the enzyme.
`Under these conditions, the sulfhydryl groups are prob(cid:173)
`ably reoxidized by molecular oxygen. Little or no stim(cid:173)
`ulatory effect is seen when compounds such as H20z,
`oxidized glutathione and cystamine, all of which can
`oxidize sulfhydryl groups, are added to enzyme that has
`
`

`
`Enzymes Which Catalyze the Initial Step in GHB Catabolism
`
`969
`
`100
`
`80
`
`r
`t=
`~ 60
`u
`
`"" ~ "" :; 40
`"
`
`f-
`
`1> e RAT
`0 A HAMSTER
`
`20
`
`4
`
`16
`
`32
`
`RATIO OF ANTIBODY TO CYTOSOL OR TO HOMOGENATE (V:V)
`
`Fig. 5. Titration of GHB dehydrogenase activity in cytosol and ho(cid:173)
`mogenate with the antibody to GHB dehydrogenase (31). The con(cid:173)
`version of [V4C]GHB to 14C02 was used as a measure of activity in
`the experiments in which homogenate was used. When cytosol was
`used, the conversion of GHB to SSA was measured spectrophoto(cid:173)
`metrically. The cytosol and homogenate used in this experiment were
`derived from the same amount of kidney. Antibody was added to the
`reaction mixture in the amounts indicated.
`
`The oxidation of GHB by the mitochondrial enzyme was
`found to be completely dependent on the presence of a(cid:173)
`ketoglutarate. The reaction was reversible. The mito(cid:173)
`chondrial enzyme also catalyzed the conversion:
`a-hydroxyglutarate + SSA ~a-ketoglutarate + GHB
`with the two products being formed in stoichiometric
`amounts. The substrate specificity is shown in Table III
`and the kinetic constants for the principal substrates in
`Table IV (20). The assumption that this enzyme is a
`transhydrogenase was confirmed when it was shown by
`gas chromatographic-mass spectroscopy that a deuterium
`on the hydroxyl-bearing carbon of one of the optical
`isomers of DL-'y-deutero-')'-hydroxybutyrate was trans(cid:173)
`ferred to the ketone-bearing carbon of a-ketoglutarate.
`This hydroxyacid-oxoacid transhydrogenase which has
`
`been found in the soluble fraction of mitochondria from
`liver, kidney and brain has been partially purified (20).
`Comparison of the Cytosolic and Mitochondrial En(cid:173)
`zymes which Oxidize GHB to SSA. With the discovery
`of the mitochondrial transhydrogenase it became appar(cid:173)
`ent that there was a dual pathway for the initial step in
`the oxidative pathway for GHB. Both cytosolic GHB
`dehydrogenase and the mitochondrial enzyme are oxi(cid:173)
`do:-eductases, but of remarkably different types. The cy(cid:173)
`tosolic enzyme is an NADP+-dependent dehydrogenase,
`whereas the mitochondrial enzyme is a pyridine nucleo(cid:173)
`tide-independent, a-ketoglutarate-dependent transhydro(cid:173)
`genase. These two enzymes, despite some striking
`differences, have one property in common: the activity
`of each of these enzymes is regulated by coupling the
`oxidation of GHB, the hydroxyacid, to the simultaneous
`reduction of an oxoacid. Though GHB dehydrogenase
`can function in the uncoupled state, conditions in the
`cytosol of most tissues (Table I) would not be favorable
`to the uncoupled reaction. The mitochondrial enzyme,
`on the other hand, has an absolute requirement for an
`oxoacid, which suggests that the metabolism of GHB by
`the mitochondrial enzyme would depend on the steady
`state levels of citric acid cycle intermediates and of a(cid:173)
`ketoglutarate in particular. The relative activities (V max
`and v) of these two enzymes in brain and kidney are
`shown in Table V. The activities of the two enzymes
`were measured under V max conditions; the rate of GHB
`oxidation (v) at average tissue GHB concentrations could
`then be calculated (Table V). How well these values
`correspond to measurements of GHB catabolism in vivo
`can be determined by examination of the half-life of
`GHB in brain. Doherty et al. (16) and Mohler et al. (15)
`have both reported a half-life (t11z) for labeled GHB in
`brain of 5 min. If one assumes that the rate of disposal
`of GHB follows first order kinetics, then from the fol-
`
`Table III. The Relative Rates of the Transhydrogenase Reaction With Other Hydroxyacids and Oxoacids (20)
`
`Hydroxy acid
`(with SSA as co-substrate)
`
`D-a-Hydroxyglutarate
`L-a-Hydroxyglutarate
`DL-13-Hydroxybutyrate
`L-13-Hydroxybutyrate
`D-13-Hydroxybutyrate
`DL-a-Hydroxybutyrate
`D-Lactate
`L-Lactate
`D-Malate
`L-Malate
`
`Relative
`Rate
`
`100
`0
`43
`45
`0
`0
`0
`1.2
`0
`0
`
`The data in this table are derived from reference (20)
`
`Oxoacid
`(with GHB as co-substrate)
`
`a-Ketoglutarate
`a-Ketoadipate
`Oxalacetate
`Pyruvate
`a-Ketobutyrate
`Acetoacetate
`13-Ketoglutarate
`13-Ketoadipate
`
`Relative
`Rate
`
`100
`24
`18.5
`8.0
`4.3
`0
`0.5
`0
`
`

`
`970
`
`Kaufman and Nelson
`
`Table IV. Kinetic Constants for the Partially Purified Hydroxyacid(cid:173)
`Ox?acid Transhydrogenase (20)
`
`Substrate
`
`"f·hydroxybutyrate
`L-(3-hydroxybutyrate
`a-Hydroxyglutarate
`a-Ketoglutarate
`Succinic semialdehyde
`
`M
`3.0 X 10- 4
`3.0 X 10-3
`4.2 X 10-4
`1.8 X 10- 4
`4.6 X 10-6
`
`lowing expression k, the first order rate constant, can be
`calculated.
`
`k = 2.3 log 2 I t112
`At a brain concentration of 2.3 nmol/g the rate of dis(cid:173)
`posal of GHB, calculated from the tt12 would be 0.32
`nmol!min/g. The combined rate of catabolism in brain
`by the two enzymes was found to be 0.18 nmol!min/g
`brain (Table V). Since the rate of disposal of GHB cal(cid:173)
`culated from the tissue half-life must include both the
`rate of catabolism of GHB and the rate of loss from brain
`by transport to the plasma, the rates calculated from the
`half-life in brain and from the combined activity of the
`two catabolic enzymes (Table V) are in reasonable
`agreement.
`Effects of Modulators of GHB Dehydrogenase In
`Vivo. One approach used to determine the magnitude of
`the contribution of GHB dehydrogenase to the metabo-
`
`lism of GHB in vivo was to give rats compounds which
`inhibit the activity of the enzyme in vitro (30). If GHB
`dehydrogenase plays a significant role in the metabolism
`of GHB in a tissue, inhibition of the enzyme in vivo
`would be expected to result in increased tissue concen(cid:173)
`trations of GHB. Two metabolic intermediates, pheny(cid:173)
`lacetate and a-ketoisocaproate, and two drugs, valproate
`and salicylate, all potent inhibitors of GHB dehydroge(cid:173)
`nase with Ki values in the range of 10- 4 to 10-5 M,
`were administered to rats. The effects of a-ketoisoca(cid:173)
`proate and phenylacetate (Table VI) and valproate and
`salicylate (table VII) on tissue contents of GHB varied
`depending on both the tissue and the compound. All of
`these compounds increased the concentration of GHB in
`one or more of the tissues assayed, in some cases by
`200%. These results are in agreement with those of Snead
`(32) who found elevated levels of GHB in brain follow(cid:173)
`ing administration of sodium valproate. Although each
`of these compounds also inhibits SSA dehydrogenase
`(30), which catalyzes the second step in the degradative
`pathway, it is very unlikely that the degree to which this
`enzyme would be inhibited is sufficient to elevate the
`concentration of SSA enough to cause the increase in
`tissue levels of GHB. The values fqr the Ki for valproate,
`for GABA transaminase (29) and for SSA dehydroge(cid:173)
`nase (29,30) are approximately 10- 2 to 10- 3M. These
`Ki values are several orders of magnitude higher than
`the K; for GHB dehydrogenase (30). From these data it
`was calculated (30) that sodium valproate at a tissue
`
`Table V. Relative Activity of the Degradative Enzymes for GHB in Brain and Kidney
`
`Enzyme
`
`GHB dehydrogenase
`
`hydroxyacid-oxoacid
`transhydrogenase
`
`GHB dehydrogenase
`
`hydroxyacid-oxoacid
`trans hydrogenase
`
`vrn<X
`nmol!min/gram
`tissue
`
`Brain
`10.1
`
`16.6
`
`Kidney
`324
`
`172
`
`Km
`(M)
`
`2.5 X 10-3
`4.5 X 1Q-4:j:
`
`3 X 10-4
`
`2.2 X 10- 3
`4.5 X 10- 4:j:
`
`3 X 10- 4
`
`Tissue
`concentration
`nmol GHB/
`gram tissue
`
`2.3
`
`28.4
`
`Calculated rate of
`oxidation of GHB at
`average tissue
`concentrations
`v = nmol!min/gram•
`
`0.01
`0.051 (coupled)
`
`0.126
`
`4.2
`19.2 (coupled)
`
`14.9
`
`b
`Vm"" • [S] h

`[S]
`= su strate concentra Ion.
`*The velocity (v) has been calculated from the equatiOn: v =
`t.
`w ere
`K., + [S]
`V max data is from reference ( 41 ), K.n data are from references (23) .and (20), tissue concentrations reference (3).
`tKm for GHB when the oxidation of GHB is coupled to the reductiOn of D-glucuronate (23).
`
`

`
`Enzymes Which Catalyze the Initial Step in GHB Catabolism
`
`971
`
`Table VI. Effects of a-Ketoisocaproate and of Phenylacetate on Tissue Levels of GHB (30)
`
`Saline
`infusion
`
`nmol GHB/g
`tissue
`
`2.6 ± 0.3 (5)
`27.8 ± 3.2 (5)
`22.2 ± 3.0 (6)
`
`Tissue
`
`Brain
`Kidney
`Muscle
`
`a-Ketoisocaproate infusion
`
`Phenylacetate infusion
`
`nmol GHB/g
`tissue
`
`3.2 ± 0.1 (3)
`55.5 ± 8.2 (4)b
`46.4 ± 9.9 (4)b
`
`Percent of
`control
`
`123
`198
`209
`
`nmol GHB/g
`tissue
`
`6.1 ± 1.0 (4)"
`18.4 ± 1.3 (4)b
`16.7 ± 7.1 (4)
`
`Percent of
`control
`
`235
`66
`75
`
`Phenylacetate (1.0 M) was given intravenously as an initial bolus of 1.5 ml followed by a constant infusion of= 2.0 ml!h for 2 h. a-Ketoisocaproate
`(0.5 M) was given intravenously as an initial bolus of 0.6 ml followed by a constant infusion of = 2.0 ml!h for 2 h. At the end of the infusion
`the animals were killed and the tissues were removed and assayed for GHB as described (30). All values are means ± SEM, numbers of animals
`are in parentheses.
`"p < 0.01
`bp < 0.05
`
`Table VII. Effects of Salicylate and of Valproate on Tissue Levels of GHB (30)
`
`Saline
`(control)
`
`nmol GHB/g
`tissue
`
`2.9 ± 0.5 (6)
`2.6 ± 0.2 (4)
`34.5 ± 5.7 (6)
`30.0 ± 4.1 (4)
`
`Tissue
`
`Brain
`
`Kidney
`
`Salicylate
`
`Valproate
`
`nmol GHB/g
`tissue
`
`5.8 ± 0.5 (4)"
`
`52.8 :t 2.5 (5)b
`
`Percent of
`control
`
`200
`
`153
`
`nmol GHB/g
`tissue
`
`Percent of
`control
`
`3.7 ± 0.2 (4)"
`
`23.7 ± 3.8 (4)
`
`142
`
`79
`
`Sodium valproate (100 mg/kg i.p.) was given 2 hr prior to decapitation; Sodium salicylate (500 mg/kg i.p.) was given 1 hr prior to decapitation.
`Tissues were removed and assayed as described (30). All values are means ± SEM, numbers of animals are in parentheses.
`"p<0.01
`bp<0.05
`
`level of 0.3 mM, a reasonable tissue concentration ex(cid:173)
`pected in therapeutic use, would produce an 85% inhi(cid:173)
`bition of GHB dehydrogenase but only a 5% inhibition
`of SSA dehydrogenase and a negligible inhibition of
`GABA transaminase. It has been reported that sodium
`valproate inhibits the conversion of SSA to GHB by an
`aldehyde reductase (EC 1.1.1.2) (29) which may be sim(cid:173)
`ilar to, or identical to, GHB dehydrogenase (EC 1.1.1.19).
`However, inhibition of GHB synthesis would not explain
`the increased tissue levels of GHB seen following
`administration of sodium valproate.
`The kinetic constants which have been determined
`for GHB dehydrogenase in vitro suggest that, in the un(cid:173)
`coupled state, the enzyme would be more likely to syn(cid:173)
`thesize GHB from SSA than to oxidize it. Indeed the
`identical enzyme, n-glucuronic reductase, is classified
`as an aldehyde reductase. It is only in the coupled state
`that the kinetic constants become favorable to the oxi(cid:173)
`dation of GHB catalyzed by this oxidoreductase under
`in vivo conditions. In kidney, D-glucuronate is likely to
`be one of the aldehydes coupled to the oxidation of GHB.
`
`In this organ mya-inositol oxygenase, the enzyme that
`synthesizes D-glucuronate, has been found to exist as a
`complex with n-glucuronate reductase (GHB dehydro(cid:173)
`genase) and is believed to transfer the newly formed n(cid:173)
`glucuronate directly to the oxidoreductase (33). Further
`proof that this oxidoreductase is probably identical to
`GHB dehydrogenase came from the finding that 95% of
`the n-glucuronate reductase activity in kidney cytosol
`and 80% of that activity in brain cytosol could be titrated
`with the polyclonal antibody to GHB dehydrogenase (31).
`What is not known is the extent to which the reaction is
`coupled in vivo and whether or not there are aldehydes
`other than D-glucuronate that can serve this role.
`To test the extent to which the enzyme is coupled
`in vivo, the effect of i.v. administration of n-glucuronate
`on the half-life of [14C]GHB in plasma has been exam(cid:173)
`ined (30). The half-life of 14C-labeled GHB in plasma
`reflects the overall rate of GHB metabolism by the body
`tissues since urinary loss of the compound is insignifi(cid:173)
`cant (34). The effect of the administration of L-gulonate,
`the product of D-glucuronate oxidation and a competitive
`
`

`
`972
`
`Kaufman and Nelson
`
`inhibitor, was also examined (30). The results shown in
`Figure 6 demonstrate that the half-life of GHB in plasma
`is shortened by D-glucuronate. The ability of o-glucu(cid:173)
`ronate to decrease the half-life of GHB in plasma indi(cid:173)
`cates that GHB dehydrogenase is not completely coupled
`in vivo. The half-life of GHB in plasma is lengthened
`by L-gulonate as would be expected from a competitive
`inhibitor of the enzyme. The effect of inhibitors of GHB
`dehydrogenase on tissue content and plasma half-life of
`GHB and the effect of D-glucuronate on the plasma half(cid:173)
`life both suggest that this enzyme may play an important
`role in the metabolism of GHB in vivo.
`NADPH is another endogenous inhibitor of GHB
`dehydrogenase (Ki = 7 x 10- 6 M) in the uncoupled
`state. The results of the experiment in which o-glucu(cid:173)
`ronate was administered to rats have shown that the re(cid:173)
`action was not completely coupled since the rate of
`metabolism could be stimulated by D-glucuronate. In the
`absence of maximal coupling, changes in the ratio of
`NADPH/NADP+ such as would occur with low ambient
`oxygen concentration might be expected to decrease the
`activity of GHB dehydrogenase and thereby increase the
`tissue concentration of GHB. The data in Table VIII
`show that in rats exposed to 5.6% oxygen for 2 hours
`the concentration of endogenous GHB is increased in
`brain from 3.0 to 10.7 nmol/gm of brain (a 3.6-fold
`increase) and in kidney from 24.6 to 57.0 nmol/gm of
`kidney (a 2.3-fold increase). This change in the redox
`
`:O.JOO
`
`~ :,000
`<
`
`;2:
`~
`-,
`
`100
`
`COMPOUND
`
`INFUSFn
`
`SALINE
`DGLUCUROr<.ATE
`LGLILONATE
`
`{6)
`{2)
`12)
`
`ll12iori 14C]GHBIM•nl
`
`-nean"!: S.EM
`60 ± 3 6
`81 = 0 5. *
`!
`40
`9
`
`*
`
`t:>·······ll L GULONATE
`
`o---o D GLUCURONATL
`
`.. "
`
`0
`
`0
`
`20
`
`40
`
`60
`
`80
`
`100
`
`120
`
`140
`
`160
`
`180
`
`200
`
`TIME {mw)
`
`Fig. 6. The effect of D·glucuronate and L·gulonate on the half-life
`(T 1/z) of GHB in plasma (30). Animals that received D-glucuronate
`received a bolus containing 240 mg followed by an infusion of 320
`mg!h for 3 h. Animals receiving L-gulonate received a bolus of 333
`mg and an infusion of 556 mg!b for 3 h. The t'/2 for the disappearance
`of [14C]GHB from plasma was determined as described. *p < 0_05;
`**p < 0.025.
`
`Table VIII. The Effect of Oxygen Concentration on Tissue Levels
`of GHB In Vivo
`
`Normal 0 2
`
`Low 0 2 (5.6%)
`
`nmol GHB/g
`tissue
`
`nmol GHB/g
`tissue
`
`Percent of
`Control
`
`Brain
`Kidney
`Muscle
`
`3.0 ± 0.15 (3)
`24.6 ± 4.2 (3)
`19.5 ± 4.2 (3)
`
`10.7 ± 0.74 (3)
`57.0 ± 13.4 (3)
`18.0 ± 2.3 (3)
`
`357*
`232
`92
`
`Animals were exposed to either room air or 5.6% oxygen for 2 hr
`before decapitation. Tissues were removed and assayed as have been
`described (3). Results are expressed as means ± SEM.
`*p = 0.0005
`
`state would also favor the synthesis of GHB by SSA
`reductase, an NADPH-dependent enzyme (35).
`Fate ofthe Carbon Skeleton ofGHB. Although GHB
`is a short chain acid and might be expected to undergo
`[3-oxidation (14), the markedly elevated plasma SSA and
`GHB concentrations in patients with GHB aciduria (17,18)
`provide evidence that [3-oxidation cannot be the major
`route of degradation. Inasmuch as the major portion of
`GHB proceeds through oxidation to SSA, the fate of the
`SSA formed either by GHB dehydrogenase or the mi(cid:173)
`tochondrial transhydrogenase must be considered. Sev(cid:173)
`eral investigators have isolated metabolic products formed
`in vivo after administration of labeled GHB (15,16,27,36-
`39). The label is distributed in citric acid cycle inter(cid:173)
`mediates and in aspartate, glutamate and GABA. Several
`reports indicate that the specific activity of GABA is
`higher than that found in glutamate. Therefore, it has
`been suggested (37,39) that the GHB skeleton may be
`converted to GABA by GABA transaminase instead of
`traversing a more circuitous route leading through the
`citric acid cycle and glutamate decarboxylase. The work
`cit

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket