Studies suggest that PE-22-28 may bind to TREK-1, a manufactured version of the naturally occurring peptide spadin. TREK-1 is localized to brain areas that regulate emotion, memory, and cognition. Researchers are looking at PE-22-28 for a variety of research applications, including within the context of depression and cognitive ability, stroke recovery aid, and in the context of neurodegenerative illnesses like Alzheimer’s.

PE-22-28 Peptide: What is it?

The naturally occurring peptide spadin has a synthetic derivative known as PE-22-28. A peptide produced from sortilin, spadin, is released by the organism. The TREK-1 receptor, which stands for TWIK-related potassium channel, is a two-pore potassium channel linked to neurogenesis and may be a target for depression. A loss of the TREK-1 receptor renders mice immune to depression, according to early research in mice. Sortilin also increases neuronal development and synaptic connectivity, increasing resistance to depression.

Curiously, spadin’s shorter analogs exhibit superior TREK-1 inhibition compared to spadin itself. This class of synthetic spadin analogs is exemplified by PE-22-28. There is data suggesting that PE-22-28 may outperform naturally occurring spadin in terms of stability, antidepressant activity, and neurogenic characteristics.

Research suggests PE-22-28 may induce neurogenesis far more quickly than any known antidepressant, with results speculated after just 4 days. Therefore, PE-22-28 may have further potential uses in learning, stroke recovery, and the fight againstneurodegenerative diseases.

Although it is most abundant in brain cells, TREK-1 may also be found in other organs and tissues such as the heart, smooth muscles, lungs, prostate, and even certain parts of the pancreas. Despite its main focus on antidepressant action, TREK-1 is involved in neuroprotection, pain perception, and anesthesia. Due to its features, academics and industry have considered TREK-1 a prominent research focus.

What is TREK-1?

Research suggests that Spadin and PE-22-28 may mostly bind to the TREK-1 receptor. TREK-1 is a multi-molecule-regulated potassium channel with two pores. As a member of the vast family of potassium channels with two pores, it plays a crucial role in controlling neuronal excitability. Specifically, TREK 1 is located in brain areas that regulate learning, memory, and emotion. This region includes the hippocampus, amygdala, and prefrontal cortex. Reducing TREK-1 function raises excitability and the probability of a depolarization event, while increasing TREK-1 activity decreases neuronal excitability. One way TREK-1 helps prevent excitotoxicity is by lowering neuronal excitability.

PE-22-28 Peptide and Depression

Based on studies conducted in animal models of depression, research has purported that PE-22-28 may alleviate behavioralpatterns congruent with depression in four days without interfering with other TREK-1 channel-controlled processes.

In research models of major depressive illness and other affective disorders, there is substantial data that the hippocampus is smaller, with lower volume. Classic antidepressants have been suggested to enhance hippocampal growth and neurogenesis in adult rats when given over an extended period. Because PE-22-28 may stimulate neurogenesis and restore this volume loss, it seems to be attacking depression at its core. Investigations purport it may aid in the discovery of some of the underlying physiological processes that are impaired in the context of depression.

PE-22-28 Peptide and Post-Stroke Depression

Following brain ischemia, many research models have post-stroke depression (PSD), which is notoriously difficult to manage with conventional methods. New data suggests that TREK-1 over-expression is a contributing factor to this disease. This increase may be inhibited or reversed in experimental mice models using SSRI antidepressants and TREK-1 blockers, such as spadin. Future studies investigating PSD may find that PE-22-28 could be successful.

PE-22-28 Peptide and Neurogenesis

Antidepressants’ capacity to enhance hippocampal neurogenesis has long been considered and researched. Based on research findings, PE-22-28 seems to be a potent peptide that may accomplish the same goal in a fraction of the time. In as little as four days, PE-22-28 has been hypothesized to enhance neurogenesis and synaptogenesis. As suggested by preliminary studies, the number of positive cells in the hippocampus for BrdU (an exogenous marker integrated into DNA and used to detect replication) appears almost doubled when PE-22-28 is presented. Regarding synaptogenesis, PE-22-28 seems to speed up the process by a factor of two.

Findings imply the upregulation of CREB after PE-22-28 may provide a further suggestion that the compound may enhance brain cell proliferation. Neuronal plasticity, memory creation, and the development of spatial memory are all linked to the transcription factor CREB, which stands for cAMP response element-binding protein. CREB seems to play an important role in neuronal development and protection. Scientists have long sought a substance that may enhance CREB for Alzheimer’s disease, as purported by research suggesting that CREB is down-regulated in Alzheimer’s disease. The potential of PE-22-28 to halt or perhaps reverse the progression of Alzheimer’s disease is a topic of intense research.

In addition to its potential for depression, the hippocampus is a crucial component of the brain region responsible for learning and memory. Research has hinted that this highly malleable structure is susceptible to damage from various insults. Depression, anxiety, and Alzheimer’s disease are just a few of the disorders that have been linked to the hippocampus. Many illnesses may be miitigated if they can regenerate better after harm. Researchers assume that because the hippocampus is involved in learning, memory, and navigation, a nootropic like PE-22-28 or another TREK-1 antagonist may be relevant for further research..

The elimination of the TREK-1 channel has been suggested to have disastrous effects in animal models. When this two-pore potassium channel was knocked out in mice models, it impaired the usual capacity of the channel to shield neurons from excitotoxicity and greatly raised the chance of seizure activity. Consequently, the fact that neither spadin nor PE-22-28 seemed to increase seizure activity was rather unexpected. The fact that spadin mice appeared to have a lower risk of experiencing generalized seizures is even more intriguing.

References

[i] J. Mazella et al., “Spadin, a sortilin-derived peptide, targeting rodent TREK-1 channels: a new concept in the antidepressant drug design,” PLoS Biol., vol. 8, no. 4, p. e1000355, Apr. 2010, doi: 10.1371/journal.pbio.1000355.

[ii] A. Djillani, M. Pietri, S. Moreno, C. Heurteaux, J. Mazella, and M. Borsotto, “Shortened Spadin Analogs Display Better TREK-1 Inhibition, In Vivo Stability and Antidepressant Activity,” Front. Pharmacol., vol. 8, Sep. 2017, doi: 10.3389/fphar.2017.00643

[iii] A. Djillani, J. Mazella, C. Heurteaux, and M. Borsotto, “Role of TREK-1 in Health and Disease, Focus on the Central Nervous System,” Front. Pharmacol., vol. 10, Apr. 2019, doi: 10.3389/fphar.2019.00379.

[iv] R. S. Duman, S. Nakagawa, and J. Malberg, “Regulation of adult neurogenesis by antidepressant treatment,” Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol., vol. 25, no. 6, pp. 836–844, Dec. 2001, doi: 10.1016/S0893- 133X(01)00358-X.

[v] J. E. Malberg and L. E. Schechter, “Increasing hippocampal neurogenesis: a novel mechanism for antidepressant drugs,” Curr. Pharm. Des., vol. 11, no. 2, pp. 145–155, 2005, doi: 10.2174/1381612053382223.

[vi] N. Katalinic, R. Lai, A. Somogyi, P. B. Mitchell, P. Glue, and C. K. Loo, “Ketamine as a new treatment for depression: a review of its efficacy and adverse effects,” Aust. N. Z. J. Psychiatry, vol. 47, no. 8, pp. 710–727, Aug. 2013, doi: 10.1177/0004867413486842.

[vii] H. Moha Ou Maati et al., “Spadin as a new antidepressant: absence of TREK-1-related side effects,” Neuropharmacology, vol. 62, no. 1, pp. 278–288, Jan. 2012, doi: 10.1016/j.neuropharm.2011.07.019.

[viii] C. Devader et al., “In vitro and in vivo regulation of synaptogenesis by the novel antidepressant spadin,” Br. J. Pharmacol., vol. 172, no. 10, pp. 2604–2617, May 2015, doi: 10.1111/bph.13083.