The long-term goal of this research is to design new compounds to inactivate GABA aminotransferase (GABA- AT), the enzyme that catabolizes the inhibitory neurotransmitter GABA, for the treatment of chemical addiction and epilepsy. Inhibition of GABA-AT, which raises GABA levels, has been shown to effectively dampen excessive neural activity without affecting basal neuronal firing. Increasing GABA levels blocks cocaine, nicotine, methamphetamine, alcohol, and heroin addiction in rats and cocaine addiction in humans without affecting the craving for food. Also, when the concentration of GABA diminishes below a threshold level in the brain, convulsions result;raising the brain GABA levels terminates the seizure. The neurochemical response to drugs of abuse is a sharp increase in dopamine levels in the nucleus accumbens, which activates the neurons responsible for pleasure and reward responses. The rise in dopamine and associated behaviors can be antagonized by an increase in the concentration of GABA. Vigabatrin (trade name SabrilTM), an irreversible inhibitor of GABA-AT, is currently marketed as a monotherapy for pediatric patients and as an adjunctive therapy for adults with refractory seizures. It was shown by positron emission tomography (PET) in primates that vigabatrin inhibits these cocaine-induced dopamine increases. The acceptance of vigabatrin for the treatment of both epilepsy and stimulant addiction, however, has been hampered by concerns about visual field defects (VFDs) in 25-50% of patients following chronic administration of large amounts of vigabatrin;the typical dose is 1-3 grams a day. The mechanism leading to the VFDs is not known;nonetheless, if the prevailing belief that VFDs arise from prolonged exposure to large doses of drug is correct, and if much lower doses of a drug can be used, there may be no untoward consequences. A new GABA-AT inactivator, (1S,3S)- 3-amino-4-difluoromethylenyl-1-cyclopentanoic acid (2), was found to be 300 times more potent than vigabatrin in modulation of the dopamine increase induced by addictive substances and in reversal of cocaine addiction in rats. Because of the potency of 2, much lower doses can be used than those with vigabatrin, which may prevent the VFDs. An important aim of this proposal is to elucidate the inactivation mechanism of 2, which will be very beneficial to future inhibitor design. Studies involving isotopic labeling of 2 and of the pyridoxal phosphate (PLP) coenzyme will be carried out. Another aim is to synthesize new inactivators of GABA-AT and study their inactivation mechanisms;one of the proposed compounds should proceed by a pathway that avoids a potential toxic by-product of vigabatrin that might produce the VFDs. Other new compounds are related to the structure of 2 to enhance potency further.
A third aim i s to determine the site of inactivator attachment on the enzyme.
A fourth aim will involve studies by collaborator Dr. Stephen Dewey on the effect of these new compounds on dopamine release in rat brains using PET and their effect on addiction in rats. The last aim is to determine the selectivity of these inactivators for GABA-AT relative to other PLP-dependent enzymes.

Public Health Relevance

The aim of this research is to design new compounds to block GABA aminotransferase, the enzyme that catalyzes the destruction of the inhibitory neurotransmitter GABA, thereby increasing the brain levels of GABA. Increasing GABA levels blocks cocaine, nicotine, methamphetamine, alcohol, and heroin addiction in rats and cocaine addiction in humans without affecting the craving for food. Also, when the concentration of GABA diminishes below a threshold level in the brain, convulsions result;raising the brain GABA levels, for example by inhibiting GABA aminotransferase, terminates the seizure.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
1R01DA030604-01
Application #
8026323
Study Section
Macromolecular Structure and Function E Study Section (MSFE)
Program Officer
Singh, Hari
Project Start
2011-01-01
Project End
2015-11-30
Budget Start
2011-01-01
Budget End
2011-11-30
Support Year
1
Fiscal Year
2011
Total Cost
$278,329
Indirect Cost
Name
Northwestern University at Chicago
Department
Chemistry
Type
Schools of Arts and Sciences
DUNS #
160079455
City
Evanston
State
IL
Country
United States
Zip Code
60201
Juncosa, Jose I; Takaya, Kenji; Le, Hoang V et al. (2018) Design and Mechanism of (S)-3-Amino-4-(difluoromethylenyl)cyclopent-1-ene-1-carboxylic Acid, a Highly Potent ?-Aminobutyric Acid Aminotransferase Inactivator for the Treatment of Addiction. J Am Chem Soc 140:2151-2164
Moschitto, Matthew J; Silverman, Richard B (2018) Synthesis of ( S)-3-Amino-4-(difluoromethylenyl)-cyclopent-1-ene-1-carboxylic Acid (OV329), a Potent Inactivator of ?-Aminobutyric Acid Aminotransferase. Org Lett 20:4589-4592
Mascarenhas, Romila; Le, Hoang V; Clevenger, Kenneth D et al. (2017) Selective Targeting by a Mechanism-Based Inactivator against Pyridoxal 5'-Phosphate-Dependent Enzymes: Mechanisms of Inactivation and Alternative Turnover. Biochemistry 56:4951-4961
Wu, Rui; Sanishvili, Ruslan; Belitsky, Boris R et al. (2017) PLP and GABA trigger GabR-mediated transcription regulation in Bacillus subtilis via external aldimine formation. Proc Natl Acad Sci U S A 114:3891-3896
Le, Hoang V; Hawker, Dustin D; Wu, Rui et al. (2015) Design and mechanism of tetrahydrothiophene-based ?-aminobutyric acid aminotransferase inactivators. J Am Chem Soc 137:4525-33
Lee, Hyunbeom; Le, Hoang V; Wu, Rui et al. (2015) Mechanism of Inactivation of GABA Aminotransferase by (E)- and (Z)-(1S,3S)-3-Amino-4-fluoromethylenyl-1-cyclopentanoic Acid. ACS Chem Biol 10:2087-98
Lee, Hyunbeom; Juncosa, Jose I; Silverman, Richard B (2015) Ornithine aminotransferase versus GABA aminotransferase: implications for the design of new anticancer drugs. Med Res Rev 35:286-305
Lee, Hyunbeom; Doud, Emma H; Wu, Rui et al. (2015) Mechanism of inactivation of ?-aminobutyric acid aminotransferase by (1S,3S)-3-amino-4-difluoromethylene-1-cyclopentanoic acid (CPP-115). J Am Chem Soc 137:2628-40
Trippier, Paul C; Jansen Labby, Kristin; Hawker, Dustin D et al. (2013) Target- and mechanism-based therapeutics for neurodegenerative diseases: strength in numbers. J Med Chem 56:3121-47
Juncosa, Jose I; Lee, Hyunbeom; Silverman, Richard B (2013) Two continuous coupled assays for ornithine-?-aminotransferase. Anal Biochem 440:145-9

Showing the most recent 10 out of 14 publications