Spinal Muscular Atrophy (SMA), a common autosomal recessive motor neuron disorder that is the leading genetic cause of infant mortality. SMA is caused by the loss of the survival motor neuron gene (SMN1). SMN2, a nearly identical copy gene, is present in all SMA patients but differs by a critical nucleotide that alters exon 7 splicing efficiency. This results in low SMN levels which are not enough to sustain motor neurons and results in a varying clinical presentation. Our previous work focused on defining the dosage requirements of SMN for normal health and restorative therapies for the severe form of the disease. Here, using newly generated mouse models we will focus on the intermediate and mild forms of SMA that have been understudied due to a lack of appropriate mouse models. We will address therapy development questions and disease mechanism in terms of SMN's cellular site of action that contributes to motor neuron dysfunction and disease pathogenesis.
In Aim 1, we will determine the timing requirements of SMN inductive therapies in intermediate and mild inducible SMA mice. This will address the potential of SMN-directed therapies to be of benefit after measureable loss has occurred and at advancing points of disease which has yet to be addressed. Here we will use a whole body Smn inductive approach to account for non-neuronal cell types that might impact disease severity.
In Aim 2, we will refine the whole body approach to focus on neuronal inductive SMN therapies using our mild SMA mouse. Specifically we will directly determine the therapeutic benefit of splice-switching anti-sense oligonucleotides (ASO) that correct SMN2 splicing. It will address whether this type of drug-candidate, when delivered after disease onset, is capable of maintaining or improving function. Importantly, this study meets an unmet milestone in the clinical development of this drug as well as other SMN inductive therapies which require CNS delivery to patients.
In Aim 3 we will compare the intrinsic electrical and synaptic properties of control and age matched SMA motor neurons from intermediate and mild SMA mice throughout their life. We postulate that altered electrical properties contribute to the cellular mechanisms underlying motor neuron dysfunction as a consequence of Smn deficiency. These studies will be performed using neonatal slice and in vitro sacral cord preparations. The in vitro preparation which we have recently developed for use in adult mice, allows intrinsic and synaptic motor neuron hyperexcitability to be assessed through intracellular recordings at time points from neonatal stages throughout adulthood. This will provide unique insights to the physiological state of SMA motor neurons throughout the course of disease. Overall, the research presented in this application will provide important information that is critical towards the development of SMN-dependent as well as independent treatment strategies for intermediate and mild forms of SMA. It also addresses an unmet need in therapy development and provides insight to the underlying cellular mechanism of disease.

Public Health Relevance

Spinal muscular atrophy (SMA) is caused by reduced levels of the survival motor neuron (SMN) protein. It is currently unknown how late in the disease process SMN inductive therapies can be beneficial in terms of either improving function or halting disease progression. This proposal focuses on 1) determining the latest time that SMN can be re-introduced after disease onset in milder forms of SMA, 2) where it is required, specifically if therapies that only increase SMN within the nervous system can correct all deficits and 3) we will compare the intrinsic electrical properties between control and age matched SMA motor neurons from intermediate and mild SMA mice throughout their life. This will allow us to correlate the physiological state of SMA motor neurons with disease progression. We believe that altered electrical properties are an early event in SMA motor neuron dysfunction. If the electrical properties are altered, identifying the timing, mechanisms and pathways involved could lead to pharmaceutical approaches that are SMN-independent and provide a complimentary approach that targets other pathways besides SMN. All of this research has important implications for therapy development.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
5R01NS060926-09
Application #
9276778
Study Section
Cell Death in Neurodegeneration Study Section (CDIN)
Program Officer
Nuckolls, Glen H
Project Start
2007-09-30
Project End
2018-05-31
Budget Start
2017-06-01
Budget End
2018-05-31
Support Year
9
Fiscal Year
2017
Total Cost
$295,953
Indirect Cost
$71,328
Name
Children's Memorial Hospital (Chicago)
Department
Type
Independent Hospitals
DUNS #
074438755
City
Chicago
State
IL
Country
United States
Zip Code
60611
Genç, Bar??; Jara, Javier H; Schultz, Megan C et al. (2016) Absence of UCHL 1 function leads to selective motor neuropathy. Ann Clin Transl Neurol 3:331-45
Singh, Natalia N; Lee, Brian M; DiDonato, Christine J et al. (2015) Mechanistic principles of antisense targets for the treatment of spinal muscular atrophy. Future Med Chem 7:1793-808
Heier, Christopher R; DiDonato, Christine J (2015) ECG in neonate mice with spinal muscular atrophy allows assessment of drug efficacy. Front Biosci (Elite Ed) 7:107-16
Keil, Jeffrey M; Seo, Joonbae; Howell, Matthew D et al. (2014) A short antisense oligonucleotide ameliorates symptoms of severe mouse models of spinal muscular atrophy. Mol Ther Nucleic Acids 3:e174
Asai, Akihiro; Chou, Pauline M; Bu, Heng-Fu et al. (2014) Dissociation of hepatic insulin resistance from susceptibility of nonalcoholic fatty liver disease induced by a high-fat and high-carbohydrate diet in mice. Am J Physiol Gastrointest Liver Physiol 306:G496-504
Gogliotti, Rocky G; Cardona, Herminio; Singh, Jasbir et al. (2013) The DcpS inhibitor RG3039 improves survival, function and motor unit pathologies in two SMA mouse models. Hum Mol Genet 22:4084-101
Sivanesan, Senthilkumar; Howell, Matthew D; Didonato, Christine J et al. (2013) Antisense oligonucleotide mediated therapy of spinal muscular atrophy. Transl Neurosci 4:
Gogliotti, Rocky G; Quinlan, Katharina A; Barlow, Courtenay B et al. (2012) Motor neuron rescue in spinal muscular atrophy mice demonstrates that sensory-motor defects are a consequence, not a cause, of motor neuron dysfunction. J Neurosci 32:3818-29
Gogliotti, Rocky G; Lutz, Cathleen; Jorgensen, Michael et al. (2011) Characterization of a commonly used mouse model of SMA reveals increased seizure susceptibility and heightened fear response in FVB/N mice. Neurobiol Dis 43:142-51
Heier, Christopher R; Hampton, Thomas G; Wang, Deli et al. (2010) Development of electrocardiogram intervals during growth of FVB/N neonate mice. BMC Physiol 10:16

Showing the most recent 10 out of 14 publications