The goal of this UH2 and UH3 is to study how exosomes can deliver siRNAs across the blood brain barrier to enter neurons and other brain cells. The immediate target is the mutant huntingtin mRNA. Huntington's disease (HD) is caused by an increase in the CAG trinucleotide repeats to ? 36 in series; it necessitates years in a high level nursing facility because of neurodegeneration first in striatum and cortex and then to other brain structures. HD patients have cognitive impairment, depression and aberrant movements. Most HD patient present by 30 to 40 years of age; a few have a juvenile onset. A rational treatment is to decrease expression of mutant huntingtin mRNA; this therapeutic can be accomplished in HD mouse models by siRNA, antisense oligonucleotides (ASO) and adeno-associated virus (AAV) with shRNAmir directed against huntingtin mRNA. However, delivery remains a pitfall to practical implementation of the therapeutics. siRNA and ASO require long-term infusion. In non-human primates, ASO administered to spinal fluid does not reach the striatum and spread of siRNA is limited in brain. Although promising, AAV-shRNA requires several injections into brain areas and the shRNAmir is unregulated. A gap in HD therapeutics can be filled by microvesicles normally extruded by cells, exosomes. Exosomes with rabies virus glycoprotein (RVG) on their surface can be injected into the blood, cross the blood brain barrier, and enter neurons and glia. RVG-exosomes can carry siRNA cargo. Delivered into the blood circulation, the exosomes deposit siRNA in neurons to engage in RNA interference. Our purpose is to develop exosomes as a therapeutic in HD. The UH2 examines the ability of RVG-exosomes carrying siRNA against huntingtin mRNA to cross the blood brain barrier to enter neurons. Localization in brain and RNAi dependent knock down will be studied. Hyper-functional siRNAs will be sought. Because exosomes are made from cytoplasm of cells, exosome mRNA, miRNA, and implaced siRNA will be identified by deep sequencing. Immune reactivity and immune-neutralization will be studied, since exosomes have potential antigens, like RVG, and will need to be administrated often. The UH3 further establishes exosome-based therapeutics, by study of reversal or prevention of neuropathology and aberrant movement in HD mouse models. Dosing of exosomes will be secured. A team of experts in HD pathogenesis, siRNA development, RNA identification and measurement, RNAi mechanisms and exosome production and brain delivery will carry out the studies. Harnessing exosomes for brain delivery is expected to form a viable therapeutic to reduce expression of mutant huntingtin in patients with HD. Patients with other genetically- based neurodegeneration will benefit.

Public Health Relevance

Huntington's disease causes memory and cognitive deficiencies, depression and abnormal movements. It is inherited as autosomal dominant. This means that each child of an affected parent has a 50% chance to inherit Huntington's disease. The disease generally starts between the ages of 30 to 40 in adults, but some patients develop the disease in adolescence. Patients with Huntington's disease often spend 10 years in costly, high level nursing facilities, because of their inability to care for themselves. Between 30,000 and 40,000 patients are affected, with many more at risk. We know the genetic cause of Huntington's disease. We know that application of RNA interference in HD animal models can prevent or ameliorate the disease. RNA interference is a method to attack and destroy mutant huntingtin gene product. However, to date, approaches for RNA interference require direct brain injection or injection into the cerebral spinal fluid. Both are suboptimal and do not have access to diseased brain cells. Exosomes provide an opportunity to treat Huntington's disease by an injection into the vein. Exosomes can be modified to cross from the blood to the brain and transport small RNAs that are used in RNA interference. Exosomes therapeutic in Huntington's disease opens up the possibility to apply them to other brain diseases.

Agency
National Institute of Health (NIH)
Institute
National Center for Advancing Translational Sciences (NCATS)
Type
Exploratory/Developmental Cooperative Agreement Phase II (UH3)
Project #
5UH3TR000888-04
Application #
9128743
Study Section
Special Emphasis Panel (ZRG1-OBT-Z (50)R)
Program Officer
Tagle, Danilo A
Project Start
2013-08-01
Project End
2018-07-31
Budget Start
2016-08-01
Budget End
2017-07-31
Support Year
4
Fiscal Year
2016
Total Cost
$980,438
Indirect Cost
$271,063
Name
University of Massachusetts Medical School Worcester
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
01655
Biscans, Annabelle; Haraszti, Reka A; Echeverria, Dimas et al. (2018) Hydrophobicity of Lipid-Conjugated siRNAs Predicts Productive Loading to Small Extracellular Vesicles. Mol Ther 26:1520-1528
Didiot, Marie-Cecile; Haraszti, Reka A; Aronin, Neil et al. (2018) Loading of Extracellular Vesicles with Hydrophobically Modified siRNAs. Methods Mol Biol 1740:199-214
Haraszti, Reka A; Coles, Andrew; Aronin, Neil et al. (2017) Loading of Extracellular Vesicles with Chemically Stabilized Hydrophobic siRNAs for the Treatment of Disease in the Central Nervous System. Bio Protoc 7:
Nikan, Mehran; Osborn, Maire F; Coles, Andrew H et al. (2017) Synthesis and Evaluation of Parenchymal Retention and Efficacy of a Metabolically Stable O-Phosphocholine-N-docosahexaenoyl-l-serine siRNA Conjugate in Mouse Brain. Bioconjug Chem 28:1758-1766
Alterman, Julia F; Coles, Andrew H; Hall, Lauren M et al. (2017) A High-throughput Assay for mRNA Silencing in Primary Cortical Neurons in vitro with Oligonucleotide Therapeutics. Bio Protoc 7:
Khvorova, Anastasia; Watts, Jonathan K (2017) The chemical evolution of oligonucleotide therapies of clinical utility. Nat Biotechnol 35:238-248
Haraszti, Reka A; Didiot, Marie-Cecile; Sapp, Ellen et al. (2016) High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. J Extracell Vesicles 5:32570
Didiot, Marie-Cécile; Hall, Lauren M; Coles, Andrew H et al. (2016) Exosome-mediated Delivery of Hydrophobically Modified siRNA for Huntingtin mRNA Silencing. Mol Ther 24:1836-1847
Coles, Andrew H; Osborn, Maire F; Alterman, Julia F et al. (2016) A High-Throughput Method for Direct Detection of Therapeutic Oligonucleotide-Induced Gene Silencing In Vivo. Nucleic Acid Ther 26:86-92
Alterman, Julia F; Hall, Lauren M; Coles, Andrew H et al. (2015) Hydrophobically Modified siRNAs Silence Huntingtin mRNA in Primary Neurons and Mouse Brain. Mol Ther Nucleic Acids 4:e266