Throughout the developing nervous system excess neurons are generated which are nonessential, or inappropriately connected, and are eliminated by programmed cell death (PCD). In the PNS, the extent of apoptosis is governed by both a limited supply of survival-promoting neurotrophic factors provided by targets of innervation, and by apoptosis-inducing competition factors secreted by ?winning neurons? that have successfully competed for these neurotrophic factors. The glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) are a family of potent growth factors that support the survival of autonomic, somatosensory and spinal motor neurons. The GFLs promote survival and growth through a common signal-transducing receptor tyrosine kinase, Ret. During this grant period we discovered that Ret interacts with p75, a member of the TNF family of death receptors, and p75 enhances GDNF-mediated Ret activation and survival. When p75 is deleted specifically in sensory neurons, approximately 20% are lost between P14 and adulthood, and these losses selectively occur in Ret+ nonpeptidergic nociceptors. These results indicate that p75 is required for the development of the nonpeptidergic nociceptor lineage by fine-tuning Ret-mediated trophic support. We also found that during PCD in sympathetic neurons of the superior cervical ganglion (SCG) Ret is restricted to a subset of degenerating neurons that rapidly undergo apoptosis. Pro-apoptotic conditions induce the association of Ret with p75, thereby enhancing the regulated intramembrane proteolysis (RIP) cleavage of p75 and activation of downstream apoptotic effectors. Deletion of p75 in Ret+ neurons, and deletion of Ret, specifically during PCD, inhibits apoptosis both in vitro and in vivo. These results indicate that Ret acts non-canonically to augment p75-mediated apoptosis. The molecular mechanisms that underlie the ability of p75 to enhance Ret signaling, and for Ret to enhance p75 mediated death, are not well understood, and are the subject of Aim 1. We also discovered recently that semaphorin 3A (Sema3A), a secreted repulsive axon guidance molecule, induces apoptosis of primary SCG neurons, and that deletion of its receptor components, Neuropilin-1 (Npn-1) and PlexinA3 (PA3), significantly reduce PCD in the SCG. Sema3A induces apoptosis via the extrinsic pathway, requiring caspase-8, as opposed to the intrinsic pathway triggered by NGF withdrawal in sympathetic neurons that requires caspase-9. The combination of apoptosis induced by neurotrophic factor deprivation and death receptor activation in the developing SCG raises the question of the extent to which caspase-8 and caspase-9 contribute to apoptosis during PCD, and which death receptors are driving this process. These questions will be the subject of Aim 2. Collectively the experiments proposed here will define the molecular mechanisms and magnitude of the role played by death receptor pathways, such as Npn-1/PA3 and p75, in PCD.

Public Health Relevance

The potent survival effects of the GFL family of growth factors have led to their development as therapeutic agents for the treatment of diseases and injuries of the nervous system. Likewise, small molecule inhibitors of p75 are currently being developed for the treatment of neurodegenerative disorders. Therefore, and understanding of the mechanisms of action of the GFLs and p75 will aid in the further development and refinement of treatment regimens based on the use of GFLs and p75 inhibitors.

Agency
National Institute of Health (NIH)
Institute
National Institute of Neurological Disorders and Stroke (NINDS)
Type
Research Project (R01)
Project #
2R01NS089585-07
Application #
9886974
Study Section
Neural Oxidative Metabolism and Death Study Section (NOMD)
Program Officer
Mamounas, Laura
Project Start
2015-05-15
Project End
2025-04-30
Budget Start
2020-05-01
Budget End
2021-04-30
Support Year
7
Fiscal Year
2020
Total Cost
Indirect Cost
Name
Indiana University-Purdue University at Indianapolis
Department
Anatomy/Cell Biology
Type
Schools of Medicine
DUNS #
603007902
City
Indianapolis
State
IN
Country
United States
Zip Code
46202
Donnelly, Christopher R; Shah, Amol A; Mistretta, Charlotte M et al. (2018) Biphasic functions for the GDNF-Ret signaling pathway in chemosensory neuron development and diversification. Proc Natl Acad Sci U S A 115:E516-E525
Shadrach, Jennifer L; Pierchala, Brian A (2018) Semaphorin3A Signaling Is Dispensable for Motor Axon Reinnervation of the Adult Neuromuscular Junction. eNeuro 5:
Donnelly, Christopher R; Gabreski, Nicole A; Suh, Esther B et al. (2018) Non-canonical Ret signaling augments p75-mediated cell death in developing sympathetic neurons. J Cell Biol 217:3237-3253
Chen, Zhijiang; Donnelly, Christopher R; Dominguez, Bertha et al. (2017) p75 Is Required for the Establishment of Postnatal Sensory Neuron Diversity by Potentiating Ret Signaling. Cell Rep 21:707-720
Wehner, A B; Milen, A M; Albin, R L et al. (2016) The p75 neurotrophin receptor augments survival signaling in the striatum of pre-symptomatic Q175(WT/HD) mice. Neuroscience 324:297-306
Gabreski, Nicole A; Vaghasia, Janki K; Novakova, Silvia S et al. (2016) Exon Skipping in the RET Gene Encodes Novel Isoforms That Differentially Regulate RET Protein Signal Transduction. J Biol Chem 291:16249-62
Wehner, Amanda B; Abdesselem, Houari; Dickendesher, Travis L et al. (2016) Semaphorin 3A is a retrograde cell death signal in developing sympathetic neurons. Development 143:1560-70
Tsui, Cynthia C; Gabreski, Nicole A; Hein, Sarah J et al. (2015) Lipid Rafts Are Physiologic Membrane Microdomains Necessary for the Morphogenic and Developmental Functions of Glial Cell Line-Derived Neurotrophic Factor In Vivo. J Neurosci 35:13233-43