SBIR-STTR Award

Resubmission: Systems for Exploring Pathogenicity in Clinical Variants of a Presynaptic Protein Involved in Epilepsy
Award last edited on: 5/12/2020

Sponsored Program
SBIR
Awarding Agency
NIH : NINDS
Total Award Amount
$224,703
Award Phase
1
Solicitation Topic Code
853
Principal Investigator
Christopher E Hopkins

Company Information

NemaMetrix LLC (AKA: NemaMetrix Inc)

44 West 7th Avenue
Eugene, OR 97403
   (541) 543-9984
   support@nemametrix.com
   www.nemametrix.com
Location: Single
Congr. District: 04
County: Lane

Phase I

Contract Number: 1R43NS108847-01A1
Start Date: 5/1/2019    Completed: 4/30/2020
Phase I year
2019
Phase I Amount
$224,703
Epilepsy is a large health problem affecting 1% of the human population. Genetic alterations in STXBP1 represents one of the most common causes of severe Early Infantile Epileptic Encephalopathy and many patients with pathogenic variations in STXBP1 are resistant to standard anti­epileptic treatment. Genome wide DNA sequencing is now being adopted in clinical practice and an increasing number of variants are identified in epilepsy­associated genes, yet the clinical interpretation of the new variants is challenging. Some of the variants are known to be either pathological or benign, yet a majority of the gene variations remain unknown for their functional consequence. A large number of Variant of Uncertain Significance (VUS) are becoming commonplace in genes for human diseases, providing a significant barrier in making diagnoses and implementing therapies. Bioinformatic approaches can provide some insight into pathogenic probability of VUS alleles, but functional studies in animal model systems are often needed to make definitive of pathogenicity assignments. The expense and long timelines of mouse models production make the use of alternative small animal models attractive. In this proposal, the C. elegans nematode is used as an alternative model capable of fast high­throughput production and screening. Human genes are installed as gene­swap replacements of the native disease­gene homologs. In preliminary work, gene­swap humanization of STXBP1 in the unc­18 locus rescued severe locomotion and behavior defects present in the gene KO animals. Further, installation of two pathogenic variants into the gene­swap locus lead to significant disruption of activity. In this proposal, a larger set of clinically­observed variants are installed as a gene­swap humanized STXBP1 locus, then function deviations are measured with various behavioral and physiological assays. In Aim 1 of proposal, a set of pathogenic variants are installed and measured capacity to exhibit detectable defects of function. In Aim 2, a set of known benign variants are installed and measured for presence of functional defects. In Aim 3, a set of VUS alleles are installed and activity is compared to the known pathogenic and benign variants. Achieving all aims provides a body of data for assessing the predictive capacity of humanized STXBP1 strains to support or refute the remaining variants for capacity to be pathogenic. In additional future phase II work, once pathogenic behavior of a variant is detected as deviant activity in the functional assays, the system can then be used to screen for compounds that lead to reversal of activity back towards wild type. As a result, success of the phase I work, create a platform for probing variant biology and drug discovery. Applied to other targets, similar discovery systems can be developed for many of the 6000+ disease associated genes of the human genome.

Public Health Relevance Statement:
Narrative Many cases of Epilepsy are caused by defects in presynaptic proteins. Doctors need to understand which of the defects cause disease. In this project, small animal models are humanized to express defective genes associated with epilepsy. The resulting humanized animal models and their analysis will be used in understanding of disease mechanisms and discovering new therapeutics.

NIH Spending Category:
Biotechnology; Brain Disorders; Epilepsy; Genetics; Human Genome; Neurodegenerative; Neurosciences

Project Terms:
Adopted; Affect; Alleles; Amino Acids; Animal Model; Animals; Antiepileptic Agents; Back; Behavior; Behavioral; Benign; Bioinformatics; Biological Assay; Biological Models; Biology; Biosensor; Caenorhabditis elegans; Clinical; clinical practice; Clustered Regularly Interspaced Short Palindromic Repeats; Code; commercialization; Data; Databases; Defect; Development; deviant; Diagnosis; Disease; DNA sequencing; drug development; drug discovery; drug testing; Early Infantile Epileptic Encephalopathy ; early onset; Epilepsy; Exhibits; Exocytosis; Frequencies; Future; Gene Expression; gene replacement; Genes; Genetic; Genetic Predisposition to Disease; genome-wide; Genomics; Health; high throughput screening; Homeostasis; Human; human disease; Human Genome; improved; insight; Laboratories; Lead; Libraries; Licensing; Locomotion; Measures; Mediating; Methods; Modeling; mouse model; Mutation; Nematoda; nervous system disorder; novel; novel therapeutics; Pathogenicity; Pathologic; Patients; personalized medicine; Pharmaceutical Preparations; Pharmacologic Substance; Phase; Phenotype; Physiological; Population; presynaptic; Probability; Production; Proteins; Reporter; Resistance; screening; Services; Site-Directed Mutagenesis; Small Business Innovation Research Grant; success; System; tau mutation; Testing; TimeLine; transcriptome sequencing; Variant; variant of unknown significance; Vesicle; Work

Phase II

Contract Number: ----------
Start Date: 00/00/00    Completed: 00/00/00
Phase II year
----
Phase II Amount
----