The development and proper function of the nervous system is critically dependent on the actions of neurotrophic factors. There are now two major families of these factors, the neurotrophins and the GDNF Family of Ligands (GFLs). At present, there are four GFLs, GDNF, Neurturin, Persephin and Artemin. These factors signal through a receptor complex composed of the Ret tyrosine kinase and a binding component (a member of a family of GPI-linked coreceptors (GFRalpha family). Experiments using in vitro cultures as well as analysis of gene-targeted mice have shown that GFLs support the survival of dopaminergic midbrain and spinal cord motor neurons as well as peripheral sensory, sympathetic, parasympathetic and enteric neurons. In this proposal, we outline experiments to study the role of Ret signaling in vivo during development as well as in adulthood. The role of GFRalpha expression in cells that lack Ret will be studied in vivo and in vitro, with particular emphasis on its role in influencing nerve regeneration and target innervation. The role of Neurturin in parasympathetic and enteric neuron differentiation and maintenance will be investigated. The physiologic roles of Artemin and Persephin will be investigated by characterizing the phenotypes of mice that lack these factors. Through this type of analysis, neuronal populations (as well as non-neuronal cells) dependent on these factors for survival as well as maintenance and proper function will be identified. Patterns of gene expression induced by Ret signaling will be assayed using microarray methodology. The focus of these studies is to understand the role of the GFLs in influencing the function of the nervous system and to determine whether abnormalities in GFL actions lead to impaired neuronal function or response to injury. This information is vital because these neurotrophic factors may be useful in reversing the neuronal deficits found in neurodegenerative diseases, peripheral neuropathies of chronic diseases such as diabetes, and gut motility syndromes.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Research Project (R01)
Project #
2R01AG013730-06
Application #
6287535
Study Section
Special Emphasis Panel (ZRG1-MDCN-7 (01))
Program Officer
Wise, Bradley C
Project Start
1996-07-01
Project End
2006-05-31
Budget Start
2001-06-01
Budget End
2002-05-31
Support Year
6
Fiscal Year
2001
Total Cost
$396,779
Indirect Cost
Name
Washington University
Department
Pathology
Type
Schools of Medicine
DUNS #
062761671
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
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
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
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