The GDNF family ligands (GFLs - GDNF, neurturin, artemin and persephin) are a family of neurotrophic factors of critical importance to neurodevelopment, and are potential therapeutic agents for many neurodegenerative diseases from Parkinson's disease to peripheral neuropathy. GFLs signal through a multimolecular complex comprised of RET (a receptor tyrosine kinase) and a specific ligand-binding coreceptor, GFRa1-4. The expression pattern of RET and the different GFRas is critical to mediate responsiveness to a particular GFL and proper neurodevelopment, but the genetic network underlying regulation of these receptors in specific neuronal subtypes remains unknown. GFLs can alter addictive behaviors through their influence on the ventral tegmental area, broadening their relevance to another critical population of midbrain dopaminergic neurons, and opening a new avenue in the treatment of substance addiction. GFLs and their receptors are expressed in the retina, and exogenous GDNF can slow retinal degeneration. Although the critical role GFLs play in peripheral nervous system development, and their promise as therapeutic agents for Parkinson's disease are firmly established, much work remains to be done to characterize the role of GFLs particularly in postnatal central nervous system development and maintenance, and to optimize and broaden their therapeutic potential into different avenues. With these goals in mind, we propose the following specific aims: (1) to identify the genetic network that regulates the expression of GFL receptors in different cell types using bioinformatics analysis of RET and the GFRa loci, together with targeted in vitro and in vivo confirmation, (2) to characterize the importance of RET signaling in midbrain dopaminergic neurons in adult animals, in regards to susceptibility to Parkinsonism-inducing toxins and modulation of addictive behaviors, using conditional removal of RET from dopaminergic neurons in transgenic mice, (3) to characterize the role of Neurturin-RET signaling in retinal development/maintenance using transgenic mice, and to investigate the utility of GFLs in a mouse model of retinal degeneration. Relevance: Neurodegenerative diseases are a tremendous burden to the public, with few effective treatments. Neurotrophic factors such as the GFLs have potential in relieving this burden, and the research in the proposal is aimed at furthering that potential.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Project (R01)
Project #
5R01AG013730-14
Application #
7643810
Study Section
Special Emphasis Panel (ZRG1-BDCN-B (92))
Program Officer
Wise, Bradley C
Project Start
1996-07-01
Project End
2011-06-30
Budget Start
2009-07-15
Budget End
2010-06-30
Support Year
14
Fiscal Year
2009
Total Cost
$563,052
Indirect Cost
Name
Washington University
Department
Pathology
Type
Schools of Medicine
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Kim, Sungsu; Maynard, Jason C; Strickland, Amy et al. (2018) Schwann cell O-GlcNAcylation promotes peripheral nerve remyelination via attenuation of the AP-1 transcription factor JUN. Proc Natl Acad Sci U S A 115:8019-8024
Essuman, Kow; Summers, Daniel W; Sasaki, Yo et al. (2018) TIR Domain Proteins Are an Ancient Family of NAD+-Consuming Enzymes. Curr Biol 28:421-430.e4
Avey, Denis; Sankararaman, Sumithra; Yim, Aldrin K Y et al. (2018) Single-Cell RNA-Seq Uncovers a Robust Transcriptional Response to Morphine by Glia. Cell Rep 24:3619-3629.e4
McGill, Bryan E; Barve, Ruteja A; Maloney, Susan E et al. (2018) Abnormal Microglia and Enhanced Inflammation-Related Gene Transcription in Mice with Conditional Deletion of Ctcf in Camk2a-Cre-Expressing Neurons. J Neurosci 38:200-219
Sasaki, Yo; Hackett, Amber R; Kim, Sungsu et al. (2018) Dysregulation of NAD+ Metabolism Induces a Schwann Cell Dedifferentiation Program. J Neurosci 38:6546-6562
Beirowski, Bogdan; Wong, Keit Men; Babetto, Elisabetta et al. (2017) mTORC1 promotes proliferation of immature Schwann cells and myelin growth of differentiated Schwann cells. Proc Natl Acad Sci U S A 114:E4261-E4270
Kim, Sungsu; Maynard, Jason C; Sasaki, Yo et al. (2016) Schwann Cell O-GlcNAc Glycosylation Is Required for Myelin Maintenance and Axon Integrity. J Neurosci 36:9633-46
Summers, Daniel W; Gibson, Daniel A; DiAntonio, Aaron et al. (2016) SARM1-specific motifs in the TIR domain enable NAD+ loss and regulate injury-induced SARM1 activation. Proc Natl Acad Sci U S A 113:E6271-E6280
Gerdts, Josiah; Summers, Daniel W; Milbrandt, Jeffrey et al. (2016) Axon Self-Destruction: New Links among SARM1, MAPKs, and NAD+ Metabolism. Neuron 89:449-60
Musiek, Erik S; Xiong, David D; Patel, Tirth et al. (2016) Nmnat1 protects neuronal function without altering phospho-tau pathology in a mouse model of tauopathy. Ann Clin Transl Neurol 3:434-42

Showing the most recent 10 out of 93 publications