Your browser doesn't support javascript.
loading
(R,S)-Ketamine metabolites (R,S)-norketamine and (2S,6S)-hydroxynorketamine increase the mammalian target of rapamycin function.
Paul, Rajib K; Singh, Nagendra S; Khadeer, Mohammed; Moaddel, Ruin; Sanghvi, Mitesh; Green, Carol E; O'Loughlin, Kathleen; Torjman, Marc C; Bernier, Michel; Wainer, Irving W.
Afiliação
  • Paul RK; From the Laboratory of Clinical Investigation (R.K.P., N.S.S., M.K., R.M., M.S., I.W.W.) and Translational Gerontology Branch (M.B.), National Institute on Aging, National Institutes of Health, Baltimore, Maryland; SRI Biosciences, SRI International, Menlo Park, California (C.E.G., K.O.); and Department of Anesthesiology, Cooper Medical School of Rowan University, Camden, New Jersey (M.C.T., I.W.W.).
Anesthesiology ; 121(1): 149-59, 2014 Jul.
Article em En | MEDLINE | ID: mdl-24936922
ABSTRACT

BACKGROUND:

Subanesthetic doses of (R,S)-ketamine are used in the treatment of neuropathic pain and depression. In the rat, the antidepressant effects of (R,S)-ketamine are associated with increased activity and function of mammalian target of rapamycin (mTOR); however, (R,S)-ketamine is extensively metabolized and the contribution of its metabolites to increased mTOR signaling is unknown.

METHODS:

Rats (n = 3 per time point) were given (R,S)-ketamine, (R,S)-norketamine, and (2S,6S)-hydroxynorketamine and their effect on the mTOR pathway determined after 20, 30, and 60 min. PC-12 pheochromocytoma cells (n = 3 per experiment) were treated with escalating concentrations of each compound and the impact on the mTOR pathway was determined.

RESULTS:

The phosphorylation of mTOR and its downstream targets was significantly increased in rat prefrontal cortex tissue by more than ~2.5-, ~25-, and ~2-fold, respectively, in response to a 60-min postadministration of (R,S)-ketamine, (R,S)-norketamine, and (2S,6S)-hydroxynorketamine (P < 0.05, ANOVA analysis). In PC-12 pheochromocytoma cells, the test compounds activated the mTOR pathway in a concentration-dependent manner, which resulted in a significantly higher expression of serine racemase with ~2-fold increases at 0.05 nM (2S,6S)-hydroxynorketamine, 10 nM (R,S)-norketamine, and 1,000 nM (R,S)-ketamine. The potency of the effect reflected antagonistic activity of the test compounds at the α7-nicotinic acetylcholine receptor.

CONCLUSIONS:

The data demonstrate that (R,S)-norketamine and (2S,6S)-hydroxynorketamine have potent pharmacological activity both in vitro and in vivo and contribute to the molecular effects produced by subanesthetic doses of (R,S)-ketamine. The results suggest that the determination of the mechanisms underlying the antidepressant and analgesic effects of (R,S)-ketamine requires a full study of the parent compound and its metabolites.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Antagonistas de Aminoácidos Excitatórios / Serina-Treonina Quinases TOR / Ketamina Limite: Animals Idioma: En Revista: Anesthesiology Ano de publicação: 2014 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Antagonistas de Aminoácidos Excitatórios / Serina-Treonina Quinases TOR / Ketamina Limite: Animals Idioma: En Revista: Anesthesiology Ano de publicação: 2014 Tipo de documento: Article