Skip to main content
U.S. flag

An official website of the United States government

A farnesyltransferase inhibitor activates lysosomes and reduces tau pathology in mice with tauopathy

Bibliographic

Year of Publication:
2019
Contact PI Name:
Kenneth S. Kosik
Contact PI Affiliation:
Neuroscience Research Institute, Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, USA
Co-Authors:
Israel Hernandez, Gabriel Luna, Jennifer N. Rauch, Surya A. Reis, Michel Giroux, Celeste M. Karch, Daniel Boctor, Youssef E. Sibih, Nadia J. Storm, Antonio Diaz, Susmita Kaushik, Cezary Zekanowski, Alexander A. Kang, Cassidy R. Hinman, et al.,
Primary Reference (PubMED ID):
Funding Source:
National Institute on Aging (NIA)
National Institute of Neurological Disorders and Stroke (NINDS)
National Science Foundation (NSF)
Information Technology Research (ITR) of the National Science Foundation (NSF)
Cohen Veterans Bioscience
The Leo and Ann Albert Charitable Trust
Tau Consortium
Postdoctoral EMBO Fellowship
Study Goal and Principal Findings:

Tau inclusions are a shared feature of many neurodegenerative diseases, among them frontotemporal dementia caused by tau mutations. Treatment approaches for these conditions include targeting posttranslational modifications of tau proteins, maintaining a steady-state amount of tau, and preventing its tendency to aggregate. We discovered a new regulatory pathway for tau degradation that operates through the farnesylated protein, Rhes, a GTPase in the Ras family. Here, we show that treatment with the farnesyltransferase inhibitor lonafarnib reduced Rhes and decreased brain atrophy, tau inclusions, tau sumoylation, and tau ubiquitination in the rTg4510 mouse model of tauopathy. In addition, lonafarnib treatment attenuated behavioral abnormalities in rTg4510 mice and reduced microgliosis in mouse brain. Direct reduction of Rhes in the rTg4510 mouse by siRNA reproduced the results observed with lonafarnib treatment. The mechanism of lonafarnib action mediated by Rhes to reduce tau pathology was shown to operate through activation of lysosomes. We finally showed in mouse brain and in human induced pluripotent stem cell-derived neurons a normal developmental increase in Rhes that was initially suppressed by tau mutations. The known safety of lonafarnib revealed in human clinical trials for cancer suggests that this drug could be repurposed for treating tauopathies.

Bibliographic Notes:
Full Author List: Israel Hernandez, Gabriel Luna1, Jennifer N. Rauch, Surya A. Reis, Michel Giroux, Celeste M. Karch, Daniel Boctor, Youssef E. Sibih, Nadia J. Storm, Antonio Diaz, Susmita Kaushik, Cezary Zekanowski, Alexander A. Kang, Cassidy R. Hinman, Vesna Cerovac, Elmer Guzman, Honjun Zhou, Stephen J. Haggarty, Alison M. Goate, Steven K. Fisher, Ana M. Cuervo, Kenneth S. Kosik.

Therapeutic Agent

Therapeutic Information:
Therapy Type:
Small Molecule
Therapeutic Agent:
Lonafarnib
Therapeutic Target:
Farnesyl Transferase
Therapy Type:
Biologic - RNA
Therapeutic Agent:
Rhes-targeted microRNA (Rhes-miR)
Therapeutic Target:
Rhes

Animal Model

Model Information:
Species:
Mouse
Model Type:
Tau
Strain/Genetic Background:
B6/FVB

Experimental Design

Is the following information reported in the study?:
Power/Sample Size Calculation
Randomized into Groups
Blinded for Treatment
Blinded for Outcome Measures
Pharmacokinetic Measures
Pharmacodynamic Measures
Toxicology Measures
ADME Measures
Biomarkers
Dose
Formulation
Route of Delivery
Duration of Treatment
Frequency of Administration
Age of Animal at the Beginning of Treatment
Age of Animal at the End of Treatment
Sex as a Biological Variable
Study Balanced for Sex as a Biological Variable
Number of Premature Deaths
Number of Excluded Animals
Statistical Plan
Genetic Background
Inclusion/Exclusion Criteria Included
Conflict of Interest
Experiment Notes

While both male and female mice were used in the experiments described, the ratio of male mice to female mice was not 1:1 for each experiment.

Outcomes

Outcome Measured
Outcome Parameters
Behavioral
Nesting Behavior
Marble Burying Test
Circling Behavior
Exploratory Activity
Histopathology
Neurofibrillary Tau Tangles
Tau Pathology
Activated Astrocytes
Activated Microglia
Biochemical
Ubiquitinated Proteins
p62/Sequestosome 1 (SQSTM1)
Rhes
phospho-Tau
Total Tau Protein
PHF Tau
Sumoylated Proteins
Autophagosomal Marker LC3-II
Autophagosomal Marker LC3-I
Sarkosyl Insoluble Tau
Microtubule Associated Protein Tau (MAPT) mRNA
Ras GTPase Family Member 2 (RASD2) mRNA
Heat Shock Proteins
Immunochemistry
phospho-Tau
Tau Protein
Glial Fibrillary Acidic Protein (GFAP)
Ionized Calcium Binding Adaptor Molecule 1 (Iba1)
Microtubule-Associated Protein 2 (MAP2)
Rhes
Immature Neurons
Synapsin
Postsynaptic Density Protein 95 (PSD95)
Microscopy
Cell Count
Cell Volume
Cell Size
Cell Viability
Cortical Volume
Hippocampal Volume
Cell Biology
Autophagic Markers
Proteolysis
PHF Tau
Pharmacodynamics
Target Engagement (Inhibition Farnesytransferase)
Omics
Whole Transcriptome Analysis
Outcomes Notes:
Pharmacodynamics: Farnesyltransferase activity was inhibited in the brains of lonafarnib-treated mice, as determined by the appearance of unfarnesylated heat shock protein HDJ-2.