Elsevier

Metabolism

Volume 55, Issue 3, March 2006, Pages 353-358
Metabolism

Metabolism of γ-hydroxybutyrate to d-2-hydroxyglutarate in mammals: further evidence for d-2-hydroxyglutarate transhydrogenase

Preliminary data were presented at the 41st Annual Meeting of the Society for the Study of Inborn Errors of Metabolism, Amsterdam, the Netherlands, 2004.
https://doi.org/10.1016/j.metabol.2005.09.009Get rights and content

Abstract

γ-Hydroxybutyratic acid (GHB), and its prodrugs 4-butyrolactone and 1,4-butanediol, represent expanding drugs of abuse, although GHB is also used therapeutically to treat narcolepsy and alcoholism. Thus, the pathway by which GHB is metabolized is of importance. The goal of the current study was to examine GHB metabolism in mice with targeted ablation of the GABA degradative enzyme succinic semialdehyde dehydrogenase (SSADH−/− mice), in whom GHB persistently accumulates, and in baboons intragastrically administered with GHB immediately and persistently. Three hypotheses concerning GHB metabolism were tested: (1) degradation via mitochondrial fatty acid β-oxidation; (2) conversion to 4,5-dihydroxyhexanoic acid (a putative condensation product of the GHB derivative succinic semialdehyde); and (3) conversion to d-2-hydroxyglutaric acid (d-2-HG) catalyzed by d-2-hydroxyglutarate transhydrogenase (a reaction previously documented only in rat). Both d-2-HG and 4,5-dihydroxyhexanoic acid were significantly increased in neural and nonneural tissue extracts derived from SSADH−/− mice. In vitro studies demonstrated the ability of 4,5-dihydroxyhexanoic acid to displace the GHB receptor ligand NCS-382 (IC50 = 38 μmol/L), although not affecting GABAB receptor binding. Blood and urine derived from baboons administered with GHB also accumulated d-2-HG, but not 4,5-dihydroxyhexanoic acid. Our results indicate that d-2-HG is a prominent GHB metabolite and provide further evidence for the existence of d-2-hydroxyglutarate transhydrogenase in different mammalian species.

Introduction

γ-Hydroxybutyratic acid (GHB), a 4-carbon hydroxy acid derived from γ-aminobutyratic acid (GABA) in brain and periphery, manifests broad pharmacological activity, including altered dopamine release and tyrosine hydroxylase activity, in addition to a number of known (and putative) receptor interactions [1]. GHB was developed as an analogue of GABA for the induction of anesthesia in humans, but early animal studies revealed unwanted side effects [2]. Renewed interest in GHB has occurred, however, in relation to its potential as a treatment for alcohol and opiate dependence and narcolepsy-associated cataplexy, as an illicit drug of abuse, and as an agent to facilitate acquaintance sexual assault [3]. Because of its capacity to induce euphoria, short-term amnesia, and sedation at high concentrations, the use of illicit GHB is expanding [4]. Unlike GHB (a controlled substance in the United States), the GHB prodrugs 4-butyrolactone and 1,4-butanediol (Fig. 1), which rapidly convert to GHB in the body, are widely accessible and uncontrolled substances, and may be potentially substituted for GHB in instances of illicit consumption [2].

Despite expanding clinical and illicit consumption, the pathways by which GHB is metabolized remain largely unexplored. Less than 2% of ingested GHB in humans is excreted unchanged in the urine, suggesting considerable metabolism [5], yet the major GHB metabolite(s) remains unknown. Walkenstein and coworkers [6] were among the first to suggest the β-oxidation of GHB (Fig. 1). In addition, urine derived from succinic semialdehyde dehyrogenase (SSADH)-deficient patients has variably shown metabolites consistent with β-oxidation, including glycolic, 3-oxo-4-hydroxybutyric, and 3,4-dihydroxybutyric acids [7]. GHB may also be metabolized to succinic semialdehyde (SSA) with stoichiometric conversion of 2-ketoglutarate to d-2-hydroxyglutaric acid (d-2-HG), in the reaction catalyzed by d-2-hydroxyglutarate transhydrogenase, an NAD(P)+-independent reaction [8] (Fig. 1). Furthermore, the presence of 4,5-dihydroxyhexanoic acid has been observed in the urine of SSADH-deficient patients, a metabolite presumably deriving from further metabolism of succinic semialdehyde (Fig. 1) [9]. At present, then, putative sequences for GHB metabolism may be represented by (1) conversion to SSA with further metabolism via the Krebs cycle (producing carbon dioxide and water, and perhaps the major metabolic pathway) [10], transamination to GABA [11], [31], [32], or conversion to 4,5-dihydroxyhexanoic acid; (2) degradation via β-oxidation; and (3) conversion to succinic semialdehyde by d-2-hydroxyglutarate transhydrogenase, with concomitant generation of d-2-HG.

GHB accumulates supraphysiologically in heritable human SSADH deficiency, a defect in the GABA degradative pathway (Fig. 1) [12], and in the corresponding gene-ablated murine model [13], [14], [15], [16]. Understanding the sequelae of GHB metabolism could have important treatment ramifications for SSADH-deficient patients and those ingesting GHB therapeutically. Accordingly, we have begun to map the mammalian metabolism of GHB. To achieve our objectives, we first evaluated GHB metabolism in SSADH-deficient (SSADH−/−) mice, followed by metabolic studies in baboons receiving short- and long-term administration of GHB [17]. In addition, the physiological significance of 4,5-dihydroxyhexanoic acid remains unknown, as it is not detected in other biological systems. Structural similarities with GHB raised the possibility that 4,5-dihydroxyhexanoic acid could compete for GHB binding. Accordingly, we tested the hypothesis that 4,5-dihydroxyhexanoic acid might be a ligand for either the high-affinity GHB or the GABAB receptors [1], [25]. The current report summarizes our findings, presented earlier in abstract form [17].

Section snippets

SSADH−/− mice

Development of SSADH−/− mice has been described [13]. Mutant (n = 6-9) and wild-type animals (n = 6-9) were age-matched (12-17 days old). For preparation of tissue extracts, mice were anesthetized with avertin and killed. Tissues (liver, brain, and kidney) were rapidly removed and frozen immediately on dry ice. Extracts were prepared by homogenization in Tris-HCl buffer (pH 8.0), rapidly deproteinized, and the extracts clarified by centrifugation followed by neutralization. Samples were stored

Free fatty acids, triglycerides, and carnitine levels in liver and serum of SSADH−/− mice

Quantification of free fatty acids in tissue extracts of SSADH−/− and SSADH+/+ mice (μmol/100 mg protein, n = 6 each genotype) revealed the following: liver, SSADH−/− 3.8 ± 1.0 (SEM) and SSADH+/+ 3.3 ± 0.7; kidney, SSADH−/− 2.3 ± 0.5 and SSADH+/+ 1.9 ± 0.2. Quantification of triglycerides (mg/100 mg protein; n = 6 except for n = 4 in SSADH−/− kidney) revealed the following: liver, SSADH−/− 20.2 ± 4.5 and SSADH+/+ 12.3 ± 1.9; kidney, SSADH−/− 64.0 ± 12.0 and SSADH+/+ 95.1 ± 13.1 (P = NS between

Discussion

Despite its expanding role as therapeutic agent and drug of abuse, surprisingly little is known about the metabolism of GHB. These metabolic sequences possess important ramifications for clinical utility and efficacy, forensic investigation, as well as the mode of action of GHB in its broad neuromodulatory activity [2]. In the current report, we sought to fill this gap in our knowledge, using as a springboard a model system (SSADH−/− mice) in which GHB accumulates to supraphysiological levels.

Acknowledgment

This work was supported in part by NS40270 (KMG), DA14919 (EMW), P20 RR17699 (MJP), DA14951 (LSQ), and a grant from the Partnership for Pediatric Epilepsy Research (including the American Epilepsy Society, the Epilepsy Foundation, Anna and Jim Fantaci, Fight Against Childhood Epilepsy and Seizures, Neurotherapy Ventures Charitable Research Fund, and Parents Against Childhood Epilepsy (KMG).

References (41)

  • C.R. Lee

    Evidence for the β-oxidation of orally administered 4-hydroxybutyrate in humans

    Biochem Med

    (1977)
  • K.M. Gibson et al.

    Succinic semialdehyde dehydrogenase deficiency associated with combined 4-hydroxybutyric and dicarboxylic acidurias: potential for clinical misdiagnosis based on urinary organic acid profiling

    J Pediatr

    (1989)
  • O.C. Snead et al.

    In vivo conversion of gamma-aminobutyric acid and 1,4-butanediol to gamma-hydroxybutyric acid in rat brain. Studies using stable isotopes

    Biochem Pharmacol

    (1989)
  • R.H. Roth et al.

    γ-Butyrolactone and γ-hydroxybutyric acid—I. Distribution and metabolism

    Biochem Pharmacol

    (1966)
  • E.A. Struys et al.

    Mutations in the d-2-hydroxyglutarate dehydrogenase gene cause d-2-hydroxyglutaric aciduria

    Am J Hum Genet

    (2005)
  • G.G. Gould et al.

    Quantitative autoradiographic analysis of the new radioligand [(3)H](2E)-(5-hydroxy-5,7,8,9-tetrahydro-6H-benzo[a][7]annulen-6-ylidene) ethanoic acid ([(3)H]NCS-382) at gamma-hydroxybutyric acid (GHB) binding sites in rat brain

    Brain Res

    (2003)
  • C.G. Wong et al.

    Gamma-hydroxybutyric acid: neurobiology and toxicology of a recreational drug

    Toxicol Rev

    (2004)
  • R. Brenneisen et al.

    Pharmacokinetics and excretion of gamma-hydroxybutyrate (GHB) in healthy subjects

    J Anal Toxicol

    (2004)
  • G.K. Brown et al.

    Urinary organic acids in succinic semialdehyde dehydrogenase deficiency: evidence of α-oxidation of 4-hydroxybutyric acid, interaction of succinic semialdehyde with pyruvate dehydrogenase and possible secondary inhibition of mitochondrial β-oxidation

    J Inherit Metab Dis

    (1987)
  • K.M. Gibson et al.

    Metabolism of [U-14C]-4-hydroxybutyric acid to intermediates of the tricarboxylic acid cycle in extracts of rat liver and kidney mitochondria

    Eur J Drug Metab Pharmacokinet

    (1989)
  • Cited by (41)

    • Determination of endogenous concentration of γ-hydroxybutyric acid (GHB) in hair through an ad hoc GC-MS analysis: A study on a wide population and influence of gender and age

      2016, Journal of Pharmaceutical and Biomedical Analysis
      Citation Excerpt :

      γ-Hydroxybutyric acid (GHB) is an endogenous compound normally present in several mammal tissues and particularly in central nervous system (CNS) [1–5].

    • Catabolism of (2E)-4-hydroxy-2-nonenal viaω- and ω-1-oxidation stimulated by ketogenic diet

      2014, Journal of Biological Chemistry
      Citation Excerpt :

      GHB was not labeled from the perfusion with [3,4-13C2]HNA, although 2-HG was M2-labeled in the perfusion with [3,4-13C2]HNA. Clearly, GHB production is coupled with the oxidation of 2-HG to 2-KG (35). In this experiment, OTHFPA-CoA catabolized from HNA/HNE was confirmed by perfusing rat livers with labeled and unlabeled OTHFPA.

    View all citing articles on Scopus
    View full text