Human glucose transporter type I-deficiency (G1D) leads to reduced brain glucose influx and neurological dysfunction principally manifested as epilepsy. Normally, most glucose is fully degraded into CO2 and water for brain energy generation via the tricarboxylic acid (TCA) cycle, which is also central to the synthesis and utilization of the neurotransmitters glutamate and GABA. Importantly, a fraction of glucose does not directly generate energy, but refills natural TCA cycle precursor loss through a reaction termed anaplerosis. Despite these long-established biochemical principles, it is unclear how most diseases that impair brain metabolism and cause seizures disrupt excitability within brain tissue (rather than in vitro), including G1D, which leads to spike-wave epilepsy. This knowledge gap about mechanisms critically limits treatment, as illustrated by anticonvulsant resistance in G1D, which is also the rule in many other neurometabolic disorders. Our laboratory and clinical long-term goal is to mechanistically understand these brain metabolism-excitability relationships in patients and mouse models to develop pharmacological and dietary therapies. The objectives of this application are to characterize hyperexcitability in a novel, robust G1D mouse model and to mitigate it by stimulating both anaplerosis and the TCA cycle with dietary substrates. Our human data and preliminary laboratory results, such as the finding of TCA cycle precursor depletion and of abnormal neocortical and thalamic excitability in G1D mice, justify investigating these mechanisms in more depth to understand epileptic hypersynchronization as a central feature of human and murine G1D. This leads to the main hypothesis that synaptic dysfunction is critical for disease pathophysiology. The proposal also includes the therapeutic consideration that even-carbon ketones, generated from common dietary fats or a ketogenic diet, can fuel the TCA cycle and ameliorate seizures in G1D, but are not anaplerotic. In contrast, our data open a new therapeutic opportunity because administered odd-carbon fat refills brain TCA cycle precursors efficiently in G1D;leading to the additional hypothesis that it restores neural functio more effectively than even-carbon fat via anaplerosis. The hypotheses will be tested in three aims: 1) Investigate the basis of cortical hyperexcitability in G1D;2) Expand this mechanistic approach to the thalamus;3) Restore brain metabolism and function via anaplerosis. The proposal is significant because its focus on metabolism-excitability relationships and anaplerosis in brain tissue using a very informative mouse model represents a shift in approach to neurometabolic diseases, where electrophysiology, 13C NMR and mass spectrometry offer a complementary understanding of mechanisms conducive to potential therapies. Particularly innovative is to combine an investigation of synaptic function in circuits or brain regions crucial for epilepsy, impaired behavior or mental retardation with the development of methodology sensitive to conscious mouse brain metabolism with broad applicability to other encephalopathies. In summary, we expect that this proposal will help define G1D as a synaptic disorder and render it amenable to excitable or metabolic target modification.

Public Health Relevance

Most epilepsies caused by mutations in genes involved in energy metabolism are intractable and can only be alleviated by life-long palliative efforts, which constitutes an important health problem. Understanding how these disorders, which are part of an expanding group of disabling diseases, are associated with seizures fulfills the NIH mission by uncovering new fundamental aspects of brain function and by facilitating the development of potential therapies aimed to restore brain energy and excitation balance.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
1R01NS077015-01A1
Application #
8373828
Study Section
Clinical Neuroplasticity and Neurotransmitters Study Section (CNNT)
Program Officer
Fureman, Brandy E
Project Start
2012-07-01
Project End
2017-06-30
Budget Start
2012-07-01
Budget End
2013-06-30
Support Year
1
Fiscal Year
2012
Total Cost
$347,630
Indirect Cost
$128,880
Name
University of Texas Sw Medical Center Dallas
Department
Neurology
Type
Schools of Medicine
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Cheshkov, Sergey; Dimitrov, Ivan E; Jakkamsetti, Vikram et al. (2017) Oxidation of [U-13 C]glucose in the human brain at 7T under steady state conditions. Magn Reson Med 78:2065-2071
Beck, Simon J; Guo, Lan; Phensy, Aarron et al. (2016) Deregulation of mitochondrial F1FO-ATP synthase via OSCP in Alzheimer's disease. Nat Commun 7:11483
Eksambe, Deepali; Agim, Nnenna; Uddin, Naseem et al. (2015) IKBKG Mutation With Incontinentia Pigmenti and Ring-Enhancing Encephalopathy. JAMA Neurol 72:1533-5
Pascual, Juan M; Ronen, Gabriel M (2015) Glucose Transporter Type I Deficiency (G1D) at 25 (1990-2015): Presumptions, Facts, and the Lives of Persons With This Rare Disease. Pediatr Neurol 53:379-93
Pascual, Juan M; Liu, Peiying; Mao, Deng et al. (2014) Triheptanoin for glucose transporter type I deficiency (G1D): modulation of human ictogenesis, cerebral metabolic rate, and cognitive indices by a food supplement. JAMA Neurol 71:1255-65
Ulate-Campos, Adriana; Fons, Carmen; Artuch, Rafael et al. (2014) Alternating hemiplegia of childhood with a de novo mutation in ATP1A3 and changes in SLC2A1 responsive to a ketogenic diet. Pediatr Neurol 50:377-9
Jeffrey, F Mark; Marin-Valencia, Isaac; Good, Levi B et al. (2013) Modeling of brain metabolism and pyruvate compartmentation using (13)C NMR in vivo: caution required. J Cereb Blood Flow Metab 33:1160-7
Marin-Valencia, Isaac; Good, Levi B; Ma, Qian et al. (2013) Heptanoate as a neural fuel: energetic and neurotransmitter precursors in normal and glucose transporter I-deficient (G1D) brain. J Cereb Blood Flow Metab 33:175-82
Marin-Valencia, Isaac; Good, Levi B; Ma, Qian et al. (2012) Glut1 deficiency (G1D): epilepsy and metabolic dysfunction in a mouse model of the most common human phenotype. Neurobiol Dis 48:92-101
Marin-Valencia, Isaac; Good, Levi B; Ma, Qian et al. (2012) Cortical metabolism in pyruvate dehydrogenase deficiency revealed by ex vivo multiplet (13)C NMR of the adult mouse brain. Neurochem Int 61:1036-43

Showing the most recent 10 out of 12 publications