Restless legs syndrome (RLS) is a chronic sleep motor disorder characterized by unpleasant sensations in the legs and an uncontrollable urge to move them for relief. Past pathophysiological studies have associated RLS to the disorder of the central dopaminergic system and iron metabolism. Family and twin studies strongly support a genetic contribution to the pathogenesis of RLS. Tremendous progress has been made recently of uncovering candidate genes linked to RLS. Three independent studies published in the last two years all pointed to the role of BTBD9 in RLS. The function of BTBD9 protein is not known. Current animal models include 6-hydroxydopamine-lesioned rodents, iron deficiency mice, and dopamine receptor 3 knockout mice. The identification of the RLS candidate genes paves the way for making genotypic model of RLS that will be more relevant in elucidating the pathophysiology of RLS and developing therapeutic treatments. The broad, long- term objective of our research is to use transgenic mice to determine: 1) the functional role of BTBD9 protein in vivo, 2) how the alteration of BTBD9 protein function could lead to RLS. The objective of this application is to characterize Btbd9 (mouse homolog of the human BTBD9 gene) knockout mice we have already made that will enable us to answer these questions. We hypothesize that BTBD9 mutations lead to alteration of brain iron metabolism and central dopaminergic system especially the striatal D2 receptor mediated indirect pathway, or diencephalic-spinal dopaminergic tracts (A11), or both. This in turn leads to enhanced unpleasant sensations, circadian dysfunction, and strong urge to move. We plan to test our hypothesis with the following Specific Aims: 1. To test the hypothesis that BTBD9 mutation disrupts dopaminergic function, we will measure animal's response to amphetamine administration in open field apparatus and examine the morphology and number of A11 neurons in hypothalamus that are known to project to spinal cord. Levels of tissue dopamine, their metabolites, and dopamine receptors will also be determined in striatum. 2. To test the hypothesis that BTBD9 mutation leads to the strong urge to move and hyperactivity, we will monitor animals'activity in home cages over several days. In addition, the mutant mice will be tested in cages with free access running wheels that are ported to a computer. These experiments will reveal whether mutant Btbd9 mice show any circadian variation of motor activity. 3. To test the hypothesis that BTBD9 mutation affects iron metabolism, we will measure striatal and nigral ferritin, transferrin and total iron in the Btbd9 mutant mice. 4. To test the hypothesis that BTBD9 mutation affects sensory system, we will measure the pain threshold using tail flick and hot plate assays, and sense of touch using Von Frey fibers. The successful completion of the above Specific Aims will help us to determine the function of BTBD9 protein in vivo and how the mutation of BTBD9 causes RLS. The results should significantly increase our understanding of the pathophysiology of RLS, which can ultimately aid the development of therapeutic treatments for RLS patients.

Public Health Relevance

The successful completion of the proposed research project will help us to determine the function of BTBD9 protein and how the mutation of BTBD9 gene causes Restless Legs Syndrome. The results should ultimately aid the development of therapeutic treatments for Restless Legs Syndrome patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Exploratory/Developmental Grants (R21)
Project #
1R21NS065273-01A1
Application #
7788666
Study Section
Clinical Neuroplasticity and Neurotransmitters Study Section (CNNT)
Program Officer
Mitler, Merrill
Project Start
2009-09-30
Project End
2011-08-31
Budget Start
2009-09-30
Budget End
2010-08-31
Support Year
1
Fiscal Year
2009
Total Cost
$219,750
Indirect Cost
Name
University of Alabama Birmingham
Department
Neurology
Type
Schools of Medicine
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Allen, Richard P; Donelson, Nathan C; Jones, Byron C et al. (2017) Animal models of RLS phenotypes. Sleep Med 31:23-28
DeAndrade, Mark P; Trongnetrpunya, Amy; Yokoi, Fumiaki et al. (2016) Electromyographic evidence in support of a knock-in mouse model of DYT1 Dystonia. Mov Disord 31:1633-1639
Yokoi, Fumiaki; Dang, Mai T; Liu, Jun et al. (2015) Decreased dopamine receptor 1 activity and impaired motor-skill transfer in Dyt1 ?GAG heterozygous knock-in mice. Behav Brain Res 279:202-10
Yokoi, Fumiaki; Chen, Huan-Xin; Dang, Mai Tu et al. (2015) Behavioral and electrophysiological characterization of Dyt1 heterozygous knockout mice. PLoS One 10:e0120916
Oleas, Janneth; Yokoi, Fumiaki; DeAndrade, Mark P et al. (2013) Engineering animal models of dystonia. Mov Disord 28:990-1000
Yokoi, Fumiaki; Cheetham, Chad C; Campbell, Susan L et al. (2013) Pre-synaptic release deficits in a DYT1 dystonia mouse model. PLoS One 8:e72491
Sciamanna, Giuseppe; Hollis, Robert; Ball, Chelsea et al. (2012) Cholinergic dysregulation produced by selective inactivation of the dystonia-associated protein torsinA. Neurobiol Dis 47:416-27
Zhang, Lin; Yokoi, Fumiaki; Parsons, Dee S et al. (2012) Alteration of striatal dopaminergic neurotransmission in a mouse model of DYT11 myoclonus-dystonia. PLoS One 7:e33669
DeAndrade, Mark P; Johnson Jr, Russell L; Unger, Erica L et al. (2012) Motor restlessness, sleep disturbances, thermal sensory alterations and elevated serum iron levels in Btbd9 mutant mice. Hum Mol Genet 21:3984-92
DeAndrade, Mark P; Zhang, Li; Doroodchi, Atbin et al. (2012) Enhanced hippocampal long-term potentiation and fear memory in Btbd9 mutant mice. PLoS One 7:e35518

Showing the most recent 10 out of 21 publications