Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Bipolar Disord. 2014 Jul 8;16(8):875–880. doi: 10.1111/bdi.12220

Valnoctamide, a non-teratogenic amide derivative of valproic acid, inhibits arachidonic acid activation in vitro by recombinant acyl-CoA synthetase-4

Hiren R Modi 1, Mireille Basselin 1, Stanley I Rapoport 1
PMCID: PMC4554599  NIHMSID: NIHMS598197  PMID: 25041123

Abstract

Objective

Valproic acid (VPA), a mood stabilizer used for treating bipolar disorder (BD), uncompetitively inhibits acylation of arachidonic acid (AA) by recombinant AA-selective acyl-CoA synthetase (Acsl)-4 at Ki of 25 mM. Inhibition may account for VPA’s ability to reduce AA turnover in brain phospholipids of unanesthetized rats at a therapeutically relevant dose. VPA is teratogenic. We now tested whether valnoctamide (VCD), a non-teratogenic amide derivative of a VPA chiral isomer, which had antimanic potency in a Phase III BD trial, also inhibits recombinant Acsl4.

Methods

Rat Acsl4-flag protein was expressed in E. coli. We used Michaelis–Menten kinetics to characterize and quantify the ability of VCD to inhibit conversion of AA to AA-CoA by recombinant Acsl4 in vitro.

Results

Acsl4-mediated activation of AA to AA-CoA by Acsl4 was inhibited uncompetitively by VCD, with Ki equal 6.38 mM.

Conclusions

VCD’s ability to uncompetitively inhibit AA activation to AA-CoA by Acsl4, at a lower Ki than VPA, suggests that, like VPA, VCD may reduce AA turnover in rat brain phospholipids. If so, VCD and other non-teratogenic Acsl4 inhibitors might be considered further for treating BD.

Keywords: acyl-CoA synthetase 4, anticonvulsant, arachidonic acid, bipolar disorder, brain, cascade, enzyme, inhibition, metabolism, Michaelis–Menten kinetics, mood stabilizer, rat, valnoctamide, valproate


Valnoctamide [valmethamide or 2-ethyl-3-methyl pentanamide (VCD)] (Fig. 1) is an over-the-counter drug available in many European countries as a sedative-hypnotic (1, 2). VCD is a chiral constitutional isomer of valpromide (VPD), the corresponding amide of valproic acid (VPA), a mood stabilizer approved by the FDA for treating bipolar disorder (BD) (25). Compared to VPA, however, VCD is significantly less teratogenic in mice (2, 6). It is not transformed into its homologous acid, valnoctic acid, or converted to VPA in vivo (4, 7, 8). It distributes better than VPA in brain (8), and is three times more potent as an anticonvulsant (3, 9).

Fig. 1.

Fig. 1

Structure of valproic acid (VPA) and valnoctamide (VCD).

VPA is effective in treating bipolar mania, but has a significant risk for birth defects and developmental delay through its epigenetic effects, particularly from inhibition of histone deacetylase (HDAC) (1012). Having a similarly acting antimanic drug that is not teratogenic would be of clinical relevance. In this regard, in a Phase III trial, VCD was more effective than placebo as an add-on to risperidone for treating bipolar mania (13). On the basis of its clinical antimanic efficacy, metabolic stability, and lack of teratogenicity, VCD has a potential to become a new BD drug.

VPA and the other FDA-approved mood stabilizers, carbamazepine, lithium, and lamotrigine, when given chronically to unanesthetized rats to produce therapeutically relevant plasma concentrations, downregulate markers of the brain arachidonic acid [(AA) 20:4n-6] cascade (14, 15): AA turnover in brain phospholipids or AA influx from plasma, expression of cyclooxygenase (COX)-2, and prostaglandin E (PGE2) concentration. Since markers of the AA cascade are upregulated in the postmortem BD brain, in association with excitotoxicity, neuroinflammation, apoptosis and synaptic loss (1618), dampening the cascade by these drugs may contribute to their efficacy in BD (14).

AA undergoes rapid deacylation-reacylation recycling within brain phospholipids (1921), and it and its products (e.g., prostaglandins, thromboxanes, leukotrienes) have multiple biological effects and participate in neurotransmission and neuroinflammation (14, 15). As part of the deacylation-reacylation cycle, AA is hydrolyzed from membrane phospholipid by AA-selective calcium-dependent cytosolic phospholipase A2 (cPLA2) IVA, which is transcriptionally downregulated in rat brain following treatment with the mood stabilizers, carbamazepine and lithium. On the other hand, VPA’s downregulation of AA turnover in rat brain has been ascribed to its ability to uncompetitively inhibit activation of AA to AA-CoA by AA-selective acyl-CoA synthetase (Acsl, E.C.6.2.1.3)-4 (2224). In uncompetitive inhibition, the inhibitor binds to the enzyme-substrate complex [ES] only and not to the free enzyme [E], while in noncompetitive inhibition the inhibitor binds to [E] or [ES]. VPA uncompetitively inhibits recombinant Acsl4 in vitro at a Ki of 25 mM (23).

In view of VPA’s ability to inhibit recombinant Acsl4 in vitro, we thought it of interest to test whether other agents that are not as teratogenic would do so well. As a first approach, we showed that a non-teratogenic constitutional isomer of VPA, propylisopropylacetic acid (PIA), also inhibited recombinant Acsl4 in vitro, but with a higher inhibition efficiency than VPA (25) and at a lower Ki, 11.4 mM. In the present study, we used similar in vitro Michaelis–Menten kinetics to test whether VCD also would inhibit recombinant Acsl4 activity.

Briefly, we found that VCD inhibited Acsl4-mediated activation of AA to AA-CoA by recombinant Acsl4 in vitro, with a lower Ki than reported for VPA and PIA (23, 25). Butyrate, an analog of VPA and also a histone deacetylase inhibitor (26), had no inhibitory action.

Materials and methods

Reagents

[1-14C]AA (50 mCi/mmol) was purchased from Moravek Biochemicals (Brea, CA, USA). Unlabeled AA, sodium butyrate, coenzyme A, and ATP were purchased from Sigma (St. Louis, MO, USA). Racemic VCD was obtained from the National Institute of Mental Health's Chemical Synthesis and Drug Supply Program (Research Triangle Park, NC, USA).

Preparation of bacterial lysate

Proteins were expressed as previously described (23, 25). Briefly, cells containing plasmids that expressed ACSL4 were grown in Terrific Broth. Protein expression was induced with 1 mM isopropyl-β-D-1-thiogalactopyranoside. Cells were pelleted and resuspended in a buffer containing 10 mM HEPES (pH 7.8) and 0.5 mM EDTA, and sonicated. Lysate aliquots were stored at −80°C for enzyme assay. Protein concentrations were determined by the Bradford method (27). As previously reported (23), we demonstrated with Western blotting and a specific anti-Flag M2 monoclonal antibody that the enzyme preparation that we studied was a single Acsl4 isoenzyme, whereas the empty control showed no immunostaining.

Acsl4 activity assay

The assay mix included 175 mM Tris-HCl pH 7.4, 8 mM MgCl2, 5 mM dithiothreitol, 10 mM ATP, 0.25 mM CoA, 0.01 mM EDTA, and 5 µM [14C]AA in 0.5 mM Triton X-100, and increasing concentrations of unlabeled AA in a total volume of 200 µl. PIA (0, 5, 10 or15 mM in ethanol), PID (10 mM in water) or MTMCD (10 mM in water), was added directly to the reaction mixture during inhibition assays. The drug controls consisted of the respective vehicle without the drug. As an additional negative control, sodium butyrate (a short-chain VPA analog) was added to the reaction mixture at 60 mM. The reaction was started by adding enzyme (1– 3 µg protein) and was measured for 5 min at 37°C (23, 25). The reaction was terminated with 1 ml Dole's Reagent (isopropanol:heptane:1M H2SO4, 80:20:2, by vol). In a preliminary experiment, the pH of reaction mixtures spiked with VPA and sodium butyrate at concentrations of 60 mM was measured using a pH meter. The pH (7.4) remained constant at these drug concentrations. Unesterified fatty acids were extracted 2–3 times with 2 ml heptane, and [14C]AA-CoA formed during the reaction was measured by scintillation counting. As a negative control, Acsl enzyme activity of the E. coli cell lysate lacking a gene coding for ACSL-Flag was measured with AA as substrate as described above. The results were corrected for blanks (samples without cell lysates added and samples analyzed in the absence of fatty acids). The negative control (empty vector) activity was compared with Acsl4 to make sure that signal to noise ratio was adequate between the test and negative control at each concentration of AA.

Analysis and statistics

Initial reaction velocity (V) was plotted against AA concentration for each VCD analogue concentration Io, and the plots were fitted by least squares to a hyperbolic Michaelis–Menten model using GraphPad Prism version 5.00 (GraphPad Software, San Diego, CA). Km (µM) and Vmax (nmol/min/mg protein) were calculated by the following equation, in which V is reaction velocity (nmol acyl-CoA formed/min/mg enzyme protein, e.g., nmol/min/mg protein) at a given AA substrate concentration, S (µM)

V=VmaxSKm+S (i)

The model in which the substrate (i.e. AA) inhibits the reaction velocity can be described as (28),

V=VmaxSKm+S(1+SKs) (ii)

A model that involves both substrate inhibition and uncompetitive inhibition by the inhibitor Io can be represented as,

V=VmaxSKm=S(1+SKs+IoKi) (iii)

where Ki is the enzyme inhibition constant.

Data were plotted as a function of inhibitor concentration Io, for VCD, and the enzyme inhibition constant (Ki) was derived from the ascending part of the plot. Lineweaver–Burke plots of 1/V versus 1/S in the presence of different inhibitor concentrations were plotted (28).

Selection of the model

To determine which inhibition model best described the data, we utilized the Akaike Information Criterion (AIC) (29),

AIC=2k2ln(L) (iv)

where k = number of parameters and L = maximized value of the likelihood function of the model. For small sample sizes, the AIC is corrected and is given as AICc (30),

AICc=N×ln(ss/N)+2K+2K(K+1)NK1 (v)

where ss is the sum of squares from the fit, N is the number of experimental observations and K is the number of parameters in the model.

The probability that the model is correct can be determined by the following equation, where Δ is the difference between AIC scores (30)

Probability=e5Δ1e5Δ (vi)

For this study with AA as a substrate, the lowest AICc was found for the uncompetitive inhibition model, as reported for VPA and PIA (23, 25, 28)

Data are presented as mean ± S.D. Linear regression analyses for obtaining Km, Vmax, Ki and other parameters were made using GraphPad Prism Version 5.0 (GraphPad Software).

Results

First, we measured conversion of AA to AA-CoA by Acsl4 in absence of VCD. As previously described, the kinetics followed a simple Michaelis–Menten model, as illustrated in Figure 2A and showed substrate inhibition (23, 25). The mean apparent Km was 4.63 ± 0.67 µM and apparent Vmax was 137.1 ± 4.80 nmol/min/mg (n = 3 independent experiments), consistent with reported values (23, 25).

Fig. 2.

Fig. 2

(A) Initial reaction velocity [(V) nmol/min/mg protein] of acyl-CoA synthetase (Acsl4) plotted against increasing arachidonic acid (AA) concentration (0 to 185 µM concentration) [S] in the presence of 0, 1.25, 2.5, or 5.0 mM VCD [I], showing substrate inhibition. (B) Typical plot of V (nmol/min/mg protein) of Acsl4 plotted against increasing AA concentration (0 to 35 µM concentration) [S] in the presence of 0, 1.25, 2.5, or 5.0 mM VCD [I]. (C) Typical Lineweaver–Burke plot of the reciprocal of reaction velocity (1/V) against inverse of substrate concentration, 1/[S] (1/[AA]), within AA concentration from 0 to 35 µM (see Results). The plot is typical of three experiments as indicated in text. Parallel plots are characteristic of uncompetitive inhibition (28).

To determine the effect of VCD on the kinetics of the reaction, VCD was added to the assay system. As illustrated in Figures 2A and 2B, VCD (1.25, 2.5, and 5.0 mM) inhibited activation of AA to AA-CoA by Acsl4. with an apparent Kiof 6.38 ± 0.63 mM (n = 3). When calculating the Lineweaver–Burke plots in Figure 2C, we considered substrate AA concentrations only in the rising phase of the V (velocity) versus [AA] curves, from 0 to 35 µM AA (Fig. 2B). At higher AA concentrations the enzyme showed substrate inhibition (Fig. 2A). The Lineweaver–Burke plot of Figure 2C showed parallel slopes, which is characteristic of uncompetitive inhibition by VCD (28). The difference between AICc values for the uncompetitive and noncompetitive enzyme inhibition models was 2.595 (Eq. v), which means that the probability that the uncompetitive model was correct was 79%, compared to 21% for the noncompetitive model (Eq. vi). In uncompetitive inhibition, the inhibitor binds to the enzyme-substrate complex [ES] only and not to free enzyme [E], while in noncompetitive inhibition the inhibitor binds to [E] or [ES]. The Lineweaver–Burke plot for a noncompetitive inhibitor shows increases in slopes with the presence of the inhibitor.

As an additional control, we measured Acsl4 activity in the presence of sodium butyrate, a 4-carbon analog of VPA that also inhibits histone deacetylase (26). As reported (23, 25), sodium butyrate did not inhibit Acsl4 activity at a concentration of 60 mM (data not shown). We also did not find any Acsl4 activity with the empty vector, consistent with our earlier observations (data not shown) (23, 25).

Discussion

We examined inhibition of the conversion of AA to AA-CoA by the non-teratogenic VPA analogue, VCD, using rat recombinant Acsl4 in vitro (23, 25). Similar to VPA and PIA, VCD inhibited Acsl4 conversion by an uncompetitive mechanism, whereas butyrate had no measurable inhibitory effect. VCD inhibited Acls4 activity with an apparent Ki of 6.38 mM, much less than the reported Ki of 25 mM for VPA and of 11.4 for PIA (23, 25). These values suggest that VCD would be more effective in vivo on an equi-concentration basis than either VPA or PIA.

As for VPA and PIA (23, 25), an uncompetitive pattern of inhibition demonstrated by Michaelis–Menten kinetics was consistent with the parallel Lineweaver–Burke plots, and it had a high probability compared with other mechanisms using the Akaike Information Criterion (AIC). Uncompetitive inhibition implies that VCD binds to the Acsl4-AA substrate complex at a different binding site than does substrate AA, and thereby causes a conformational change that reduces enzyme activity and AA activation to AA-CoA (28).

A therapeutically relevant dose of VPA (200 mg/kg, i.p.) in rats produced a brain VPA concentration of 1.0 to 1.5 mM (31, 32), about 10-fold less than VPA’s Ki when inhibiting recombinant Acsl4 in vitro. It was suggested that, if VPA inhibition of rat brain AA turnover in vivo is due to its inhibition of brain Acsl4, then VPA must be concentrated 10-fold at the enzyme site (23). This is possible, since studies indicate that VPA can accumulate, via a short-chain fatty acid transporter, in cellular mitochondria, microsomes and other organelles in which Acsl4 is found (3338). A similar consideration would apply to VCD, whose brain concentration at its therapeutically relevant dose (20 mg/kg, i.v.) in rats reaches 0.6 mmol/kg (8), also about 10-fold less than its in vitro Ki with recombinant Acsl4. Additionally, for each drug, the in vivo kinetic inhibition constant for Acsl4 may be less than the measured in vitro value, as the latter depends on bath conditions like pH, temperature, salt and ATP concentrations, and on the absence of transporter proteins that are found in the intact brain (39). VCD is retained in the body (in dogs) for a relatively long period of time, and plasma levels of its corresponding acid, valnoctic acid, are much lower than those of VPA (4).

VCD has been shown to be more effective than placebo as an add-on to risperidone in manic patients (13). VPA, approved by the FDA in 1995 for treating bipolar mania, is a popular alternative to lithium for BD. Chronically given VPA to rats reduces AA turnover and downregulates markers of the brain AA cascade as do the other FDA approved mood stabilizers (14, 15, 40). Since markers of the cascade are upregulated in the postmortem BD brain, in association with evidence of excitotoxicity, neuroinflammation, apoptosis and synaptic loss (16, 17), dampening of the brain AA cascade by VPA and other mood stabilizers may contribute to their efficacy in BD (14, 40). Several mechanisms of action for VPA have been proposed such as inhibiting voltage gated Na+ channels, potentiation of GABAergic activity by GABA-transaminase inhibition, Inhibition of T-type Ca2+ channels, suppression of NMDA-mediated excitation, HDAC inhibition and reduced AA turnover. Because it inhibits the chromatin-modifying enzyme, HDAC VPA also is teratogenic (10, 11). As such, it poses a significant fetal risk for pregnant women taking the drug (12), thus justifying the need for a non-teratogenic yet equipotent mood stabilizer that may act by the same mechanism as VPA. Identifying a pharmacological brain target of VPA with regard to BD could lead to rational development of effective VPA-like compounds with fewer side effects, including teratogenicity, such as VCD.

It remains to be determined experimentally whether the observed inhibition by VCD of AA to AA-CoA conversion by recombinant Acsl4 in vitro corresponds to VCD’s ability to reduce AA turnover within rat brain phospholipids in vivo, as we found with VPA. Showing this would suggest that VCD or similar non-teratogenic inhibitors of Acsl4 in vitro be considered further for drug trials in BD patients (13) and, furthermore, that the recombinant Acsl4 inhibition assay could be used for screening for new VPA-like mood stabilizers.

Acknowledgements

This work was supported by the Intramural Research Program of the National Institute on Aging, National Institute of Health. We thank the Chemical Synthesis and Drug Supply Program of the National Institute of Mental Health (Research Triangle Park, NC, USA) for supplying us with valnoctamide.

Footnotes

Disclosures

The authors of this paper do not have any commercial associations that might pose a conflict of interest in connection with this manuscript.

References

  • 1.Bialer M, Yagen B. Valproic acid: second generation. Neurotherapeut. 2007;4:130–137. doi: 10.1016/j.nurt.2006.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kaufmann D, Yagen B, Minert A, et al. Evaluation of the antiallodynic, teratogenic and pharmacokinetic profile of stereoisomers of valnoctamide, an amide derivative of a chiral isomer of valproic acid. Neuropharmacology. 2010;58:1228–1236. doi: 10.1016/j.neuropharm.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • 3.Barel S, Yagen B, Schurig V, et al. Stereoselective pharmacokinetic analysis of valnoctamide in healthy subjects and in patients with epilepsy. Clin Pharmacol Ther. 1997;61:442–449. doi: 10.1016/S0009-9236(97)90194-6. [DOI] [PubMed] [Google Scholar]
  • 4.Haj-Yehia A, Bialer M. Pharmacokinetics of a valpromide isomer, valnoctamide, in dogs. J Pharm Sci. 1988;77:831–834. doi: 10.1002/jps.2600771003. [DOI] [PubMed] [Google Scholar]
  • 5.Spiegelstein O, Yagen B, Bennett GD, Finnell RH, Blotnik S, Bialer M. Stereoselective pharmacokinetic analysis of valnoctamide, a CNS-active chiral amide analogue of valproic acid, in dogs, rats, and mice. Ther Drug Monit. 2000;22:574–581. doi: 10.1097/00007691-200010000-00012. [DOI] [PubMed] [Google Scholar]
  • 6.Radatz M, Ehlers K, Yagen B, Bialer M, Nau H. Valnoctamide, valpromide and valnoctic acid are much less teratogenic in mice than valproic acid. Epilepsy Res. 1998;30:41–48. doi: 10.1016/s0920-1211(97)00095-8. [DOI] [PubMed] [Google Scholar]
  • 7.Bialer M, Haj-Yehia A, Barzaghi N, Pisani F, Perucca E. Pharmacokinetics of a valpromide isomer, valnoctamide, in healthy subjects. Eur J Clin Pharmacol. 1990;38:289–291. doi: 10.1007/BF00315032. [DOI] [PubMed] [Google Scholar]
  • 8.Blotnik S, Bergman F, Bialer M. Disposition of valpromide, valproic acid, and valnoctamide in the brain, liver, plasma, and urine of rats. Drug Metab Dispos. 1996;24:560–564. [PubMed] [Google Scholar]
  • 9.Haj-Yehia A, Bialer M. Structure-pharmacokinetic relationships in a series of valpromide derivatives with antiepileptic activity. Pharm Res. 1989;6:683–689. doi: 10.1023/a:1015934321764. [DOI] [PubMed] [Google Scholar]
  • 10.Brown NA, Farmer PB, Coakley M. Valproic acid teratogenicity: demonstration that the biochemical mechanism differs from that of valproate hepatotoxicity. Biochem Soc Trans. 1985;13:75–77. doi: 10.1042/bst0130075. [DOI] [PubMed] [Google Scholar]
  • 11.Phiel CJ, Zhang F, Huang EY, Guenther MG, Lazar MA, Klein PS. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem. 2001;276:36734–36741. doi: 10.1074/jbc.M101287200. [DOI] [PubMed] [Google Scholar]
  • 12.Samren EB, van Duijn CM, Koch S, et al. Maternal use of antiepileptic drugs and the risk of major congenital malformations: a joint European prospective study of human teratogenesis associated with maternal epilepsy. Epilepsia. 1997;38:981–990. doi: 10.1111/j.1528-1157.1997.tb01480.x. [DOI] [PubMed] [Google Scholar]
  • 13.Bersudsky Y, Applebaum J, Gaiduk Y, et al. Valnoctamide as a valproate substitute with low teratogenic potential in mania: a double-blind, controlled, add-on clinical trial. Bipolar Disord. 2010;12:376–382. doi: 10.1111/j.1399-5618.2010.00828.x. [DOI] [PubMed] [Google Scholar]
  • 14.Kim HW, Rapoport SI, Rao JS. Altered expression of apoptotic factors and synaptic markers in postmortem brain from bipolar disorder patients. Neurobiology Disease. 2010;37:596–603. doi: 10.1016/j.nbd.2009.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shimizu T, Wolfe LS. Arachidonic acid cascade and signal transduction. J Neurochem. 1990;55:1–15. doi: 10.1111/j.1471-4159.1990.tb08813.x. [DOI] [PubMed] [Google Scholar]
  • 16.Kim HW, Rapoport SI, Rao JS. Altered expressions of apoptotic factors and synaptic markers in postmortem brain from bipolar disorder patients. Neurobiology Disease. 2010;37:596–603. doi: 10.1016/j.nbd.2009.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kim HW, Rapoport SI, Rao JS. Altered arachidonic acid cascade enzymes in postmortem brain from bipolar disorder patients. Mol Psychiatry. 2011;16:419–428. doi: 10.1038/mp.2009.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rao JS, Kellom M, Kim HW, Rapoport SI, Reese EA. Neuroinflammation and synaptic loss. Neurochem Res. 2012;37:903–910. doi: 10.1007/s11064-012-0708-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lands WEM, Crawford CG. Enzymes of membrane phospholipid metabolism in animals. In: Martonosi AN, editor. The Enzymes of Biological Membranes: Biosynthesis of Cell Components. New York: Plenum; 1976. pp. 3–85. [Google Scholar]
  • 20.Sun GY, MacQuarrie RA. Deacylation-reacylation of arachidonoyl groups in cerebral phospholipids. Ann N Y Acad Sci. 1989;559:37–55. doi: 10.1111/j.1749-6632.1989.tb22597.x. [DOI] [PubMed] [Google Scholar]
  • 21.Rapoport SI. Arachidonic acid and the brain. J Nutr. 2008;138:2515–2520. doi: 10.1093/jn/138.12.2515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bazinet RP, Weis MT, Rapoport SI, Rosenberger TA. Valproic acid selectively inhibits conversion of arachidonic acid to arachidonoyl-CoA by brain microsomal long-chain fatty acyl-CoA synthetases: relevance to bipolar disorder. Psychopharmacology. 2006;184:122–129. doi: 10.1007/s00213-005-0272-4. [DOI] [PubMed] [Google Scholar]
  • 23.Shimshoni JA, Basselin M, Li LO, Coleman RA, Rapoport SI, Modi HR. Valproate uncompetitively inhibits arachidonic acid acylation by rat acyl-CoA synthetase 4: relevance to valproate's efficacy against bipolar disorder. Biochim Biophys Acta. 2011;1811:163–169. doi: 10.1016/j.bbalip.2010.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Soupene E, Kuypers FA. Mammalian long-chain acyl-CoA synthetases. Experiment Biol Med. 2008;233:507–521. doi: 10.3181/0710-MR-287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Modi HR, Basselin M, Taha AY, et al. Propylisopropylacetic acid (PIA), a constitutional isomer of valproic acid, uncompetitively inhibits arachidonic acid acylation by rat acyl-CoA synthetase 4: a potential drug for bipolar disorder. Biochim Biophys Acta. 2013;1831:880–886. doi: 10.1016/j.bbalip.2013.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Eyal S, Yagen B, Shimshoni J, Bialer M. Histone deacetylases inhibition and tumor cells cytotoxicity by CNS-active VPA constitutional isomers and derivatives. Biochem Pharmacol. 2005;69:1501–1508. doi: 10.1016/j.bcp.2005.02.012. [DOI] [PubMed] [Google Scholar]
  • 27.Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248–254. doi: 10.1016/0003-2697(76)90527-3. [DOI] [PubMed] [Google Scholar]
  • 28.Palmer T. Understanding Enzymes. New York: John Wiley; 1985. [Google Scholar]
  • 29.Akaike H. A new look at statistical model identification. IEEE Trans Autom Control. 1974;19:716–723. [Google Scholar]
  • 30.Motulsky CA. Fitting Models to Biological Data using Linear and Nonlinear Regression: A Practical Guide to Curve Fitting. New York: Oxford University Press'; 2004. pp. 143–148. [Google Scholar]
  • 31.Deutsch J, Rapoport SI, Rosenberger TA. Valproyl-CoA and esterified valproic acid are not found in brains of rats treated with valproic acid, but the brain concentrations of CoA and acetyl-CoA are altered. Neurochem Res. 2003;28:861–866. doi: 10.1023/a:1023267224819. [DOI] [PubMed] [Google Scholar]
  • 32.Loscher W, Fisher JE, Nau H, Honack D. Valproic acid in amygdala-kindled rats: alterations in anticonvulsant efficacy, adverse effects and drug and metabolite levels in various brain regions during chronic treatment. J Pharmacol Exp Ther. 1989;250:1067–1078. [PubMed] [Google Scholar]
  • 33.Scism JL, Powers KM, Artru AA, Lewis L, Shen DD. Probenecid-inhibitable efflux transport of valproic acid in the brain parenchymal cells of rabbits: a microdialysis study. Brain Res. 2000;884:77–86. doi: 10.1016/s0006-8993(00)02893-6. [DOI] [PubMed] [Google Scholar]
  • 34.Rajaonarison JF, Lacarelle B, Catalin J, Placidi M, Rahmani R. 3'-azido-3'-deoxythymidine drug interactions. Screening for inhibitors in human liver microsomes. Drug Metab Dispos. 1992;20:578–584. [PubMed] [Google Scholar]
  • 35.Terlouw SA, Tanriseven O, Russel FG, Masereeuw R. Metabolite anion carriers mediate the uptake of the anionic drug fluorescein in renal cortical mitochondria. J Pharmacol Exp Ther. 2000;292:968–973. [PubMed] [Google Scholar]
  • 36.Cao Y, Murphy KJ, McIntyre TM, Zimmerman GA, Prescott SM. Expression of fatty acid-CoA ligase 4 during development and in brain. FEBS Lett. 2000;467:263–267. doi: 10.1016/s0014-5793(00)01159-5. [DOI] [PubMed] [Google Scholar]
  • 37.Cao Y, Traer E, Zimmerman GA, McIntyre TM, Prescott SM. Cloning, expression, and chromosomal localization of human long-chain fatty acid-CoA ligase 4 (FACL4) Genomics. 1998;49:327–330. doi: 10.1006/geno.1998.5268. [DOI] [PubMed] [Google Scholar]
  • 38.Meloni I, Muscettola M, Raynaud M, et al. FACL4, encoding fatty acid-CoA ligase 4, is mutated in nonspecific X-linked mental retardation. Nature Genet. 2002;30:436–440. doi: 10.1038/ng857. [DOI] [PubMed] [Google Scholar]
  • 39.Knudsen J, Neergaard TB, Gaigg B, Jensen MV, Hansen JK. Role of acyl-CoA binding protein in acyl-CoA metabolism and acyl-CoA-mediated cell signaling. J Nutr. 2000;130:S294–S298. doi: 10.1093/jn/130.2.294S. [DOI] [PubMed] [Google Scholar]
  • 40.Rao JS, Rapoport SI. Mood-stabilizers target the brain arachidonic acid cascade. Curr Mol Pharmacol. 2009;2:207–214. doi: 10.2174/1874467210902020207. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES