Peripheral neuropathic pain results from maladaptive changes in the central nervous system that are initiated by abnormal activity of injured sensory neurons. Increasing evidence indicates that neuroplasticity in the spinal cord depends on glial-neuronal interactions. In neuropathic pain, glial cells play a critical role in both the development and maintenance of hypersensitivity. Our data suggest that the neuropeptide precursor protein VGF (non-acronymic) and the peptides derived from it may represent novel injury-induced glial activators. We have shown that 1) VGF is rapidly and robustly upregulated in sensory neurons at the onset of nerve injury, and 2) VGF-derived peptides activate spinal microglia. VGF peptides also evoke thermal hyperalgesia, and mechanical and cold allodynia, and potentiate the activity of dorsal horn neurons. These results strongly implicate VGF peptides as sensory neuron signals that initiate microglial activation after nerve injury and participate in spinal neuroplasticity. Furthermore, VGF remains upregulated for the duration of the behavioral hypersensitivity, indicating that VGF peptides may participate both in the initiation of neuropathic pain and also in its maintenance. Our long-term research goal is to delineate the role that VGF-derived neuropeptides play in conditions of chronic pain. Our objective in this application is to determine how VGF peptides drive the spinal neuroplasticity that leads to neuropathic pain, and to establish whether they are required for its maintenance. The central hypothesis of this proposal is that VGF peptides serve as key mediators of the transition from acute to chronic pain.
In Specific Aim 1, we will determine the role of VGF peptides in the spinal neuroplasticity that leads to neuropathic pain. We hypothesize that VGF peptides are required for microglial activation and the development of nerve injury-induced hypersensitivity, and that VGF ablation or immunoneutralization of VGF peptides will prevent these events.
In Specific Aim 2, we will determine the contribution of VGF to the maintenance of the neuropathic pain state. We hypothesize that the levels and/or processing of VGF peptides in spinal cord and CSF are altered for the duration of neuropathic pain and that neuropathic pain will be abolished by VGF ablation.
In Specific Aim 3, we will delineate the mechanisms responsible for VGF regulation of spinal neuroplasticity. We hypothesize that VGF peptides regulate the neuromodulatory activity of microglia that drives maladaptive neuroplasticity in dorsal horn neurons. Characterization of biologically active VGF-derived peptides, which may additionally establish novel CSF biomarkers of the neuropathic state, the VGF signaling system, and the peptide receptors that mediate VGF actions, has the potential to identify new therapeutic approaches to control spinal neuroplasticity, providing insight into the development and progression of neuropathic pain.

Public Health Relevance

Increased effort to understand what causes pain has motivated a multi-decade global research effort to identify new molecular players that communicate the perception of pain to the brain. The proposed research will investigate the activity of a novel spinal pain signaling system, that of the neuropeptide precursor protein VGF (non-acronymic). We have recently shown that under conditions of chronic pain, this protein increases in sensory neurons. We hypothesize that this protein contributes to injury signals that mediate the development of chronic pain. Understanding how VGF drives pain signaling may lead to new approaches for diagnosing and treating chronic pain conditions.

Agency
National Institute of Health (NIH)
Institute
National Institute of Dental & Craniofacial Research (NIDCR)
Type
Research Project (R01)
Project #
5R01DE021996-04
Application #
8704122
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Wan, Jason
Project Start
2011-08-01
Project End
2016-07-31
Budget Start
2014-08-01
Budget End
2015-07-31
Support Year
4
Fiscal Year
2014
Total Cost
Indirect Cost
Name
University of Minnesota Twin Cities
Department
Veterinary Sciences
Type
Overall Medical
DUNS #
City
Minneapolis
State
MN
Country
United States
Zip Code
55455
Jiang, Cheng; Lin, Wei-Jye; Salton, Stephen R (2018) Role of a VGF/BDNF/TrkB Autoregulatory Feedback Loop in Rapid-Acting Antidepressant Efficacy. J Mol Neurosci :
Skorput, Alexander G J; Zhang, Xijing; Waataja, Jonathan J et al. (2018) Involvement of the VGF-derived peptide TLQP-62 in nerve injury-induced hypersensitivity and spinal neuroplasticity. Pain 159:1802-1813
Stephens, Samuel B; Edwards, Robert J; Sadahiro, Masato et al. (2017) The Prohormone VGF Regulates ? Cell Function via Insulin Secretory Granule Biogenesis. Cell Rep 20:2480-2489
Choi, Soon Gang; Wang, Qian; Jia, Jingjing et al. (2016) Characterization of Gonadotrope Secretoproteome Identifies Neurosecretory Protein VGF-derived Peptide Suppression of Follicle-stimulating Hormone Gene Expression. J Biol Chem 291:21322-21334
Guedon, Jean-Marc G; Wu, Shaogen; Zheng, Xuexing et al. (2015) Current gene therapy using viral vectors for chronic pain. Mol Pain 11:27
Sadahiro, Masato; Erickson, Connor; Lin, Wei-Jye et al. (2015) Role of VGF-derived carboxy-terminal peptides in energy balance and reproduction: analysis of ""humanized"" knockin mice expressing full-length or truncated VGF. Endocrinology 156:1724-38
Lin, Wei-Jye; Jiang, Cheng; Sadahiro, Masato et al. (2015) VGF and Its C-Terminal Peptide TLQP-62 Regulate Memory Formation in Hippocampus via a BDNF-TrkB-Dependent Mechanism. J Neurosci 35:10343-56
Fairbanks, Carolyn A; Peterson, Cristina D; Speltz, Rebecca H et al. (2014) The VGF-derived peptide TLQP-21 contributes to inflammatory and nerve injury-induced hypersensitivity. Pain 155:1229-37
Cero, Cheryl; Vostrikov, Vitaly V; Verardi, Raffaello et al. (2014) The TLQP-21 peptide activates the G-protein-coupled receptor C3aR1 via a folding-upon-binding mechanism. Structure 22:1744-1753
Lin, Wei-Jye; Salton, Stephen R (2013) The regulated secretory pathway and human disease: insights from gene variants and single nucleotide polymorphisms. Front Endocrinol (Lausanne) 4:96