Spinal and Bulbar Muscular Atrophy (SBMA) is a progressive neuromuscular disease affecting the proximal spinal and bulbar motor neurons and the skeletal muscles. To date, there is no effective cure for this devastating disease. SBMA arises from the expansion of a polymorphic CAG trinucleotide repeat in the Androgen Receptor (AR) gene, resulting in an expansion of glutamines in the AR protein. Polyglutamine expansion renders the mutant AR protein toxic, resulting in the formation of mutant protein aggregates and cell death. The overarching goal of this project is to better understand the cellular and molecular mechanisms underlying the pathogenesis of SBMA with the hope that such insight will lead to the development of better therapeutics. In order to reach this goal, we began by identifying factors that could potentially modulate the SBMA disease condition in vivo. The preliminary data presented in this application clearly show that Nemo-Like Kinase (NLK), an evolutionarily conserved serine/threonine kinase, is a key factor that could potentially modulate the SBMA disease condition. In this proposal, we will test the idea that NLK can modulate SBMA pathogenesis via regulating the posttranslational modification or expression of the mutant AR. To investigate this idea, we propose the following three specific aims.
In Aim 1, we will determine whether NLK can modulate SBMA-related phenotypes using mouse as a model system. We will utilize several SBMA mouse models that are characterized by both neuromuscular pathology and behavioral deficits. Specifically, (1) we will first test whether constitutive loss of one functional Nlk allele (50% NLK reduction) suppresses the neuromuscular phenotypes of SBMA. (2) We will also evaluate the role of NLK in SBMA in a tissue-specific manner by using Nlk conditional knockout mice. We will focus on motor neurons and skeletal muscle, as they are known SBMA- affected tissues.
In Aim 2, we will identify the molecular mechanism by which NLK regulates the SBMA disease condition. We will determine how NLK kinase activity is responsible for mediating the neuromuscular phenotypes of SBMA. The role of NLK in (1) AR posttranslational modification and (2) AR protein degradation/clearance will be determined and their contribution to SBMA toxicity will be characterized.
In Aim 3, we will determine the role of the peptidyl-prolyl isomerase Pin1 in SBMA pathology. We will test whether Pin1 acts downstream of the NLK-induced effects on mutant AR to modulate toxicity in SBMA. To do this, we will utilize cell culture approaches and Drosophila and mouse genetics. We believe that the knowledge gained from the studies proposed in this application will advance our basic understanding of the cellular and molecular mechanisms underlying SBMA and will suggest new therapeutic interventions aimed at reducing the burden of SBMA and other neuromuscular diseases.

Public Health Relevance

The burden of neuromuscular diseases is large, yet no effective therapeutics exists for a majority of these devastating disorders. The studies outlined i this proposal will allow us to determine key pathogenic pathways underlying the neuromuscular disease Spinal and Bulbar Muscular Atrophy, with the hope that this study may lead to the development of novel therapeutics.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
3R01NS088321-04S1
Application #
9767438
Study Section
Cellular and Molecular Biology of Neurodegeneration Study Section (CMND)
Program Officer
Gubitz, Amelie
Project Start
2015-09-01
Project End
2020-08-31
Budget Start
2018-09-01
Budget End
2019-08-31
Support Year
4
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Yale University
Department
Genetics
Type
Schools of Medicine
DUNS #
043207562
City
New Haven
State
CT
Country
United States
Zip Code
Driessen, Terri M; Lee, Paul J; Lim, Janghoo (2018) Molecular pathway analysis towards understanding tissue vulnerability in spinocerebellar ataxia type 1. Elife 7:
Khalid, Madiha; Driessen, Terri M; Lee, Jong Seo et al. (2018) Association of CACNA1C with bipolar disorder among the Pakistani population. Gene 664:119-126
Tejwani, Leon; Lim, Janghoo (2017) Hunting for the mutant without the MAP(K). Cell Res 27:1403-1404
Todd, Tiffany W; Kokubu, Hiroshi; Miranda, Helen C et al. (2015) Nemo-like kinase is a novel regulator of spinal and bulbar muscular atrophy. Elife 4:e08493