Viral vectors encoding endomorphins and serine histogranin attenuate neuropathic pain symptoms after spinal cord injury in rats

Mol Pain. 2015 Jan 7:11:2. doi: 10.1186/1744-8069-11-2.

Abstract

Background: The treatment of spinal cord injury (SCI)-induced neuropathic pain presents a challenging healthcare problem. The lack of available robust pharmacological treatments underscores the need for novel therapeutic methods and approaches. Due to the complex character of neuropathic pain following SCI, therapies targeting multiple mechanisms may be a better choice for obtaining sufficient long-term pain relief. Previous studies in our lab showed analgesic effects using combinations of an NMDA antagonist peptide [Ser1]histogranin (SHG), and the mu-opioid peptides endomorphins (EMs), in several pain models. As an alternative to drug therapy, this study evaluated the analgesic potential of these peptides when delivered via gene therapy.

Results: Lentiviruses encoding SHG and EM-1 and EM-2 were intraspinally injected, either singly or in combination, into rats with clip compression SCI 2 weeks following injury. Treated animals showed significant reduction in mechanical and thermal hypersensitivity, compared to control groups injected with GFP vector only. The antinociceptive effects of individually injected components were modest, but the combination of EMs and SHG produced robust and sustained antinociception. The onset of the analgesic effects was observed between 1-5 weeks post-injection and sustained without decrement for at least 7 weeks. No adverse effects on locomotor function were observed. The involvement of SHG and EMs in the observed antinociception was confirmed by pharmacologic inhibition using intrathecal injection of either the opioid antagonist naloxone or an anti-SHG antibody. Immunohistochemical analysis showed the presence of SHG and EMs in the spinal cord of treated animals, and immunodot-blot analysis of CSF confirmed the presence of these peptides in injected animals. In a separate group of rats, delayed injection of viral vectors was performed in order to mimic a more likely clinical scenario. Comparable and sustained antinociceptive effects were observed in these animals using the SHG-EMs combination vectors compared to the group with early intervention.

Conclusions: Findings from this study support the potential for direct gene therapy to provide a robust and sustained alleviation of chronic neuropathic pain following SCI. The combination strategy utilizing potent mu-opioid peptides with a naturally-derived NMDA antagonist may produce additive or synergistic analgesic effects without the tolerance development for long-term management of persistent pain.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Cell Line, Tumor
  • Disease Models, Animal
  • Genetic Vectors / physiology
  • Humans
  • Hyperalgesia / drug therapy
  • Lentivirus / genetics
  • Male
  • Neuralgia / drug therapy*
  • Neuralgia / etiology*
  • Neuroblastoma / pathology
  • Neuropeptides / biosynthesis
  • Neuropeptides / therapeutic use
  • Opioid Peptides / biosynthesis
  • Opioid Peptides / genetics
  • Opioid Peptides / therapeutic use*
  • Pain Measurement
  • Pain Threshold / drug effects
  • Proteins / genetics
  • Proteins / therapeutic use*
  • Rats
  • Rats, Sprague-Dawley
  • Recovery of Function / drug effects
  • Spinal Cord Injuries / complications*

Substances

  • EM-1-NHNH2
  • EM-2-NHNH2
  • Neuropeptides
  • Opioid Peptides
  • Proteins
  • histogranin