Mitochondrial-Protective Effects of R-Phenibut after Experimental Traumatic Brain Injury

Joint Authors

Kupats, Einars
Stelfa, Gundega
Zvejniece, Baiba
Grinberga, Solveiga
Vavers, Edijs
Makrecka-Kuka, Marina
Svalbe, Baiba
Zvejniece, Liga
Dambrova, Maija

Source

Oxidative Medicine and Cellular Longevity

Issue

Vol. 2020, Issue 2020 (31 Dec. 2020), pp.1-12, 12 p.

Publisher

Hindawi Publishing Corporation

Publication Date

2020-11-21

Country of Publication

Egypt

No. of Pages

12

Main Subjects

Biology

Abstract EN

Altered neuronal Ca2+ homeostasis and mitochondrial dysfunction play a central role in the pathogenesis of traumatic brain injury (TBI).

R-Phenibut ((3R)-phenyl-4-aminobutyric acid) is an antagonist of the α2δ subunit of voltage-dependent calcium channels (VDCC) and an agonist of gamma-aminobutyric acid B (GABA-B) receptors.

The aim of this study was to evaluate the potential therapeutic effects of R-phenibut following the lateral fluid percussion injury (latFPI) model of TBI in mice and the impact of R- and S-phenibut on mitochondrial functionality in vitro.

By determining the bioavailability of R-phenibut in the mouse brain tissue and plasma, we found that R-phenibut (50 mg/kg) reached the brain tissue 15 min after intraperitoneal (i.p.) and peroral (p.o.) injections.

The maximal concentration of R-phenibut in the brain tissues was 0.6 μg/g and 0.2 μg/g tissue after i.p.

and p.o.

administration, respectively.

Male Swiss-Webster mice received i.p.

injections of R-phenibut at doses of 10 or 50 mg/kg 2 h after TBI and then once daily for 7 days.

R-Phenibut treatment at the dose of 50 mg/kg significantly ameliorated functional deficits after TBI on postinjury days 1, 4, and 7.

Seven days after TBI, the number of Nissl-stained dark neurons (N-DNs) and interleukin-1beta (IL-1β) expression in the cerebral neocortex in the area of cortical impact were reduced.

Moreover, the addition of R- and S-phenibut at a concentration of 0.5 μg/ml inhibited calcium-induced mitochondrial swelling in the brain homogenate and prevented anoxia-reoxygenation-induced increases in mitochondrial H2O2 production and the H2O2/O ratio.

Taken together, these results suggest that R-phenibut could serve as a neuroprotective agent and promising drug candidate for treating TBI.

American Psychological Association (APA)

Kupats, Einars& Stelfa, Gundega& Zvejniece, Baiba& Grinberga, Solveiga& Vavers, Edijs& Makrecka-Kuka, Marina…[et al.]. 2020. Mitochondrial-Protective Effects of R-Phenibut after Experimental Traumatic Brain Injury. Oxidative Medicine and Cellular Longevity،Vol. 2020, no. 2020, pp.1-12.
https://search.emarefa.net/detail/BIM-1206031

Modern Language Association (MLA)

Kupats, Einars…[et al.]. Mitochondrial-Protective Effects of R-Phenibut after Experimental Traumatic Brain Injury. Oxidative Medicine and Cellular Longevity No. 2020 (2020), pp.1-12.
https://search.emarefa.net/detail/BIM-1206031

American Medical Association (AMA)

Kupats, Einars& Stelfa, Gundega& Zvejniece, Baiba& Grinberga, Solveiga& Vavers, Edijs& Makrecka-Kuka, Marina…[et al.]. Mitochondrial-Protective Effects of R-Phenibut after Experimental Traumatic Brain Injury. Oxidative Medicine and Cellular Longevity. 2020. Vol. 2020, no. 2020, pp.1-12.
https://search.emarefa.net/detail/BIM-1206031

Data Type

Journal Articles

Language

English

Notes

Includes bibliographical references

Record ID

BIM-1206031