Mammalian target of rapamycin in spinal cord neurons mediates hypersensitivity induced by peripheral inflammation

EN Gregory, S Codeluppi, JA Gregory, J Steinauer… - Neuroscience, 2010 - Elsevier
EN Gregory, S Codeluppi, JA Gregory, J Steinauer, CI Svensson
Neuroscience, 2010Elsevier
mTOR, the mammalian target of rapamycin, is a serine–threonine kinase known to regulate
cell proliferation and growth. mTOR has also been implicated in neuronal synaptic plasticity
as well as in pain transmission in models of chemically induced and neuropathic pain. To
date, the role of mTOR as a modulator of inflammatory pain has not been examined. In this
study, we investigated the role of mTOR in Sprague–Dawley rats using the carrageenan
model of inflammatory pain. mRNA of Ras homolog enriched in brain (Rheb), a GTPase that …
mTOR, the mammalian target of rapamycin, is a serine–threonine kinase known to regulate cell proliferation and growth. mTOR has also been implicated in neuronal synaptic plasticity as well as in pain transmission in models of chemically induced and neuropathic pain. To date, the role of mTOR as a modulator of inflammatory pain has not been examined. In this study, we investigated the role of mTOR in Sprague–Dawley rats using the carrageenan model of inflammatory pain. mRNA of Ras homolog enriched in brain (Rheb), a GTPase that positively regulates mTOR activation, was significantly increased 2 h following carrageenan injection. Four hours after induction of inflammation phosphorylation (p) of p70S6 kinase (S6K), ribosomal protein S6 (S6) and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) was increased, indicating mTOR activation. Inhibition of spinal mTOR with intrathecal (i.t.) injection of rapamycin (0.1–3 μg) led to a dose-dependent decrease in carrageenan-induced thermal hyperalgesia and a reduction of mechanical allodynia. In vitro studies confirmed rapamycin inhibition of the mTOR pathway. Carrageenan-induced activation of the mTOR pathway in rats was localized predominantly to dorsal horn neurons in the superficial lamina. Taken together, these data show that the mTOR pathway is activated in dorsal horn neurons during inflammatory pain, and that inhibition of spinal mTOR attenuates inflammation-induced thermal and tactile hypersensitivity. Hence, our study indicates that spinal mTOR is an important regulator of spinal sensitization and suggests that targeting mTOR may provide a new avenue for pain therapy.
Elsevier