Abstract

The nerve growth factor (NGF) and its mimetics, which have neuroprotective and neuroregenerative properties, are attractive candidates for developing new drugs for brain injury therapy. A dipeptide mimetic of NGF loop 4, bis(N-succinyl-L-glutamyl-L-lysine) hexamethylenediamide (GK-2), developed at the Zakusov Research Institute of Pharmacology, has the NGF-like ability to activate TrkA receptors, but unlike NGF, GK-2 activates mainly the PI3K/AKT pathway associated with neuroprotection and has no effect on the MAPK cascade associated with hyperalgesia, the main side effect of NGF. That GK-2 possesses neuroprotective activity has been observed in various models of cerebral ischemia. GK-2 was found to statistically significantly reduce the cerebral infarct volume in experimental stroke, even at treatment onset 24 h after injury. This suggests that GK-2 possesses neuroregenerative properties, which may be associated with the activation of neurogenesis and/or synaptogenesis. We studied the effect of GK-2 on neurogenesis and synaptogenesis in experimental ischemic stroke caused by transient occlusion of the middle cerebral artery in rats. GK-2 was administered 6 or 24 h after surgery and then once a day for 7 days. One day after the last administration, proliferative activity in the hippocampus and striatum of the affected hemisphere was assessed using Ki67 and synaptogenesis in the striatum was evaluated using synaptophysin and PSD-95. Ki67 immunoreactivity, both in the striatum and in the hippocampus of the ischemic rats, was found to have dropped by approximately 30% compared to that in the sham-operated controls. Synaptic markers - synaptophysin and PSD-95 - were also statistically significantly reduced, by 14 and 29%, respectively. GK-2 in both administration schedules completely restored the level of Ki67 immunoreactivity in the hippocampus and promoted its increase in the striatum. In addition, GK-2 restored the level of the postsynaptic marker PSD-95, with the therapeutic effect amounting to 70% at the start of its administration after 6 h, and promoted restoration of the level of this marker at the start of administration 24 h after an experimental stroke. GK-2 had no effect on the synaptophysin level. These findings suggest that the neurotrophin mimetic GK-2, which mainly activates one of the main Trk receptor signaling pathways PI3K/ AKT, has a stimulating effect on neurogenesis (and, probably, gliogenesis) and synaptogenesis in experimental cerebral ischemia. This effect may explain the protective effect observed at the start of dipeptide administration 24 h after stroke simulation.

Highlights

  • IntroductionThe development of pathogenic treatments for an ischemic stroke after reperfusion remains a challenge in modern medicine

  • The development of pathogenic treatments for an ischemic stroke after reperfusion remains a challenge in modern medicine.The nerve growth factor (NGF) is an attractive neuroprotective agent capable of affecting the main mechanisms of ischemic neuronal injury: glutamate toxicity mediating excessive calcium entry into the cell [1]; oxidative stress [2]

  • To shed light on this issue, we studied the effect of GK-2 on neurogenesis and synaptogenesis in an experimental ischemic stroke using antibodies to the Ki67 proliferation marker and synaptogenesis markers

Read more

Summary

Introduction

The development of pathogenic treatments for an ischemic stroke after reperfusion remains a challenge in modern medicine. The nerve growth factor (NGF) is an attractive neuroprotective agent capable of affecting the main mechanisms of ischemic neuronal injury: glutamate toxicity mediating excessive calcium entry into the cell [1]; oxidative stress [2]. NGF was experimentally proven to be involved in neurogenesis in the adult brains of rodents. The efficacy of intracerebral and intranasal administration of NGF has been proven in various cerebral ischemia models in rodents. It is extremely important that the neurotrophin remains active upon delayed administration – 24 h after a simulated stroke [6, 10]. The disadvantages of the full-length NGF protein may be obviated through the development of related low-molecular-weight mimetics with a selective pharmacological activity [12]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call